1
|
Cleary JA, Kumar A, Craft S, Deep G. Neuron-derived extracellular vesicles as a liquid biopsy for brain insulin dysregulation in Alzheimer's disease and related disorders. Alzheimers Dement 2025; 21:e14497. [PMID: 39822132 PMCID: PMC11848159 DOI: 10.1002/alz.14497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 11/24/2024] [Accepted: 12/01/2024] [Indexed: 01/19/2025]
Abstract
Extracellular vesicles (EVs) have emerged as novel blood-based biomarkers for various pathologies. The development of methods to enrich cell-specific EVs from biofluids has enabled us to monitor difficult-to-access organs, such as the brain, in real time without disrupting their function, thus serving as liquid biopsy. Burgeoning evidence indicates that the contents of neuron-derived EVs (NDEs) in blood reveal dynamic alterations that occur during neurodegenerative pathogenesis, including Alzheimer's disease (AD), reflecting a disease-specific molecular signature. Among these AD-specific molecular changes is brain insulin-signaling dysregulation, which cannot be assessed clinically in a living patient and remains an unexplained co-occurrence during AD pathogenesis. This review is focused on delineating how NDEs in the blood may begin to close the gap between identifying molecular changes associated with brain insulin dysregulation reliably in living patients and its connection to AD. This approach could lead to the identification of novel early and less-invasive diagnostic molecular biomarkers for AD. HIGHLIGHTS: Neuron-derived extracellular vesicles (NDEs) could be isolated from peripheral blood. NDEs in blood reflect the molecular signature of Alzheimer's disease (AD). Brain insulin-signaling dysregulation plays a critical role in AD. NDEs in blood could predict brain insulin-signaling dysregulation. NDEs offer novel early and less-invasive diagnostic biomarkers for AD.
Collapse
Affiliation(s)
- Jacob Alexander Cleary
- Department of Internal Medicine‐Gerontology and Geriatric MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Ashish Kumar
- Department of Internal Medicine‐Gerontology and Geriatric MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Suzanne Craft
- Department of Internal Medicine‐Gerontology and Geriatric MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
- Sticht Center for Healthy Aging and Alzheimer's PreventionWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Gagan Deep
- Department of Internal Medicine‐Gerontology and Geriatric MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
- Sticht Center for Healthy Aging and Alzheimer's PreventionWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
- Atrium Health Wake Forest Baptist Comprehensive Cancer CenterWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| |
Collapse
|
2
|
Lu H, Li Z, Zhu L, Xu P, Wang H, Li Y, Zhao W. Fabrication and Temporal Dependency Osteogenic Regulation of Dual-Scale Hierarchical Microstructures on Medical Metal Surface. Adv Healthc Mater 2024; 13:e2402369. [PMID: 39175381 DOI: 10.1002/adhm.202402369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 08/09/2024] [Indexed: 08/24/2024]
Abstract
The structural characteristics at the interface of bone implants can guide biological regulation. In this study, a dual-scale hierarchical microstructure is proposed and customized using hybrid machining to achieve temporal dependency osteogenic regulation. It is observed that osteoblasts induced by dual-scale hierarchical structure exhibit adequate protrusion development and rapid cell attachment through the modulation of mechanical forces in the cell growth environment, and further promot the upregulation of the cell membrane receptor PDGFR-α, which is related to cell proliferation. Afterward, transcriptomic analysis reveals that during the differentiation stage, the DSH structure regulates cellular signaling cascades primarily through integrin adhesion mechanisms and then accelerates osteogenic differentiation by activating the TGF-β pathway and cAMP signaling pathway. Furthermore, the calcium nodules are preferentially deposited within the lower honeycomb-like channels, thereby endowing the proposed dual-scale hierarchical structure with the potential to induce oriented deposition and improve the long-term stability of the implant.
Collapse
Affiliation(s)
- Hao Lu
- School of Mechanical Engineering and Automation, Northeastern University, Shenyang, 110819, China
| | - Zhijun Li
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China
| | - Lida Zhu
- School of Mechanical Engineering and Automation, Northeastern University, Shenyang, 110819, China
| | - Peihua Xu
- School of Mechanical Engineering and Automation, Northeastern University, Shenyang, 110819, China
| | - Hai Wang
- Shenyang Lebuy Vacuum Tech. Co., Ltd, Shenyang, Liaoning, China
| | - Yonghao Li
- Shenyang Lebuy Vacuum Tech. Co., Ltd, Shenyang, Liaoning, China
| | - Weidong Zhao
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China
| |
Collapse
|
3
|
Liu C, Gao J, Cheng Y, Zhang S, Fu C. Homologous-adhering/targeting cell membrane- and cell-mediated delivery systems: a cancer-catch-cancer strategy in cancer therapy. Regen Biomater 2024; 12:rbae135. [PMID: 39811105 PMCID: PMC11729729 DOI: 10.1093/rb/rbae135] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 10/09/2024] [Accepted: 11/06/2024] [Indexed: 01/16/2025] Open
Abstract
Low tumor enrichment remains a serious and urgent problem for drug delivery in cancer therapy. Accurate targeting of tumor sites is still a critical aim in cancer therapy. Though there have been a variety of delivery strategies to improve the tumor targeting and enrichment, biological barriers still cause most delivered guests to fail or be excreted before they work. Recently, cell membrane-based systems have attracted a huge amount of attention due to their advantages such as easy access, good biocompatibility and immune escape, which contribute to their biomimetic structures and specific surface proteins. Furthermore, cancer cell membrane-based delivery systems are referred to as homologous-targeting function in which they exhibit significantly high adhesion and internalization to homologous-type tumor sites or cells even though the exact mechanism is not entirely revealed. Here, we summarize the sources and characterizations of cancer cell membrane systems, including reconstructed single or hybrid membrane-based nano-/microcarriers, as well as engineered cancer cells. Additionally, advanced applications of these cancer cell membrane systems in cancer therapy are categorized and summarized according to the components of membranes. The potential factors related to homologous targeting of cancer cell membrane-based systems are also discussed. By discussing the applications, challenges and opportunities, we expect the cancer cell membrane-based homologous-targeting systems to have a far-reaching development in preclinic or clinics.
Collapse
Affiliation(s)
- Chenguang Liu
- Zhejiang Provincial Engineering Research Center of New Technologies and Applications for Targeted Therapy of Major Diseases, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, P. R. China
| | - Jingjie Gao
- Zhejiang Provincial Engineering Research Center of New Technologies and Applications for Targeted Therapy of Major Diseases, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, P. R. China
| | - Yuying Cheng
- Zhejiang Provincial Engineering Research Center of New Technologies and Applications for Targeted Therapy of Major Diseases, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, P. R. China
| | - Shanshan Zhang
- Department of Orthopaedics Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P. R. China
| | - Caiyun Fu
- Zhejiang Provincial Engineering Research Center of New Technologies and Applications for Targeted Therapy of Major Diseases, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, P. R. China
| |
Collapse
|
4
|
Mondaza-Hernandez JL, Hindi N, Fernandez-Serra A, Ramos R, Gonzalez-Cámpora R, Gómez-Mateo MC, Martinez-Trufero J, Lavernia J, Lopez-Pousa A, Laínez N, Martinez-Garcia J, Valverde C, Vaz-Salgado MÁ, Garcia-Plaza G, Marin-Borrero I, Carrillo-Garcia J, Martin-Ruiz M, Romero P, Gutierrez A, López-Guerrero JA, Moura DS, Martin-Broto J. Exploratory analysis of immunomodulatory factors identifies L1CAM as a prognostic marker in alveolar soft-part sarcoma. Ther Adv Med Oncol 2024; 16:17588359241293951. [PMID: 39502403 PMCID: PMC11536517 DOI: 10.1177/17588359241293951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 10/09/2024] [Indexed: 11/08/2024] Open
Abstract
Background Alveolar soft-part sarcoma (ASPS) is a rare tumor driven by the ASPSCR1-TFE3 fusion protein, with a propensity for metastasis. Prognostic factors remain poorly understood, and traditional chemotherapies are largely ineffective. Recent interest lies in immune checkpoint inhibitors (ICIs), yet predictive biomarkers for treatment response are lacking. Previous studies have shown promising results with ICIs in ASPS, indicating a need for further investigation into biomarkers associated with immune response. Objectives To identify prognostic biomarkers in ASPS and to explore the role of immune-related markers, particularly L1CAM, in predicting patient outcomes. Design A retrospective cohort study of 19 ASPS patients registered in the GEIS database. The study involved the collection of clinical and histopathological data, followed by an analysis of immune markers and gene expression profiles to identify potential prognostic indicators. Methods Clinical and histopathological data were retrospectively collected from the GEIS-26 study cohort of 19 ASPS patients. Immunohistochemistry was performed to evaluate immune markers programmed death-1 ligand (PD-L1), programmed death-1, FAS, FASL, CD8, CD3, and CD4. An HTG ImmunOncology panel was conducted on formalin-fixed paraffin-embedded samples to explore gene expression. Effects of differentially expressed genes on survival were explored by Kaplan-Meier. Results PD-L1 positivity was widely observed (63%) in tumors, and CD8+ lymphocytic infiltration was common. High CD8 density correlated with greater overall survival (OS) while not statistically significant. No associations were found for other immune markers. L1CAM was identified as differentially expressed in patients with low CD8 infiltration and correlated negatively with OS. Conclusion High L1CAM expression correlated with poorer OS, highlighting its potential as a prognostic marker and therapeutic target in ASPS. Immunomodulatory interventions may hold promise, as evidenced by PD-L1 expression and CD8+ infiltration. Further research, including larger cohorts and international collaborations, is needed to validate these findings and explore therapeutic strategies targeting L1CAM in ASPS.
Collapse
Affiliation(s)
- José L. Mondaza-Hernandez
- Health Research Institute Fundacion Jimenez Diaz, Universidad Autonoma de Madrid, Madrid, Spain
- University Hospital General de Villalba, Madrid, Spain
| | - Nadia Hindi
- Health Research Institute Fundacion Jimenez Diaz, Universidad Autonoma de Madrid, Madrid, Spain
- University Hospital General de Villalba, Madrid, Spain
- Medical Oncology Department, Fundación Jimenez Diaz University Hospital, Madrid, Spain
| | | | - Rafael Ramos
- Pathology Department, University Hospital Son Espases, Mallorca, Spain
| | | | | | | | - Javier Lavernia
- Medical Oncology Department, Fundación Instituto Valenciano de Oncologia, Valencia, Spain
| | | | - Nuria Laínez
- Department of Medical Oncology, Complejo Hospitalario de Navarra Pamplona, Spain
| | | | - Claudia Valverde
- Medical Oncology Department, Vall d’Hebron University Hospital, Barcelona, Spain
| | - María Ángeles Vaz-Salgado
- Medical Oncology Department, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria, CIBERONC, Madrid, Spain
| | - Gabriel Garcia-Plaza
- Department of Surgery, Hospital Universitario Insular, Las Palmas de Gran Canaria, Spain
| | - Isabel Marin-Borrero
- Health Research Institute Fundacion Jimenez Diaz, Universidad Autonoma de Madrid, Madrid, Spain
| | - Jaime Carrillo-Garcia
- Health Research Institute Fundacion Jimenez Diaz, Universidad Autonoma de Madrid, Madrid, Spain
- University Hospital General de Villalba, Madrid, Spain
| | - Marta Martin-Ruiz
- Health Research Institute Fundacion Jimenez Diaz, Universidad Autonoma de Madrid, Madrid, Spain
- University Hospital General de Villalba, Madrid, Spain
| | - Pablo Romero
- Health Research Institute Fundacion Jimenez Diaz, Universidad Autonoma de Madrid, Madrid, Spain
- University Hospital General de Villalba, Madrid, Spain
| | - Antonio Gutierrez
- Hematology Department, Hospital Son Espases/IdISBa, Palma de Mallorca, Spain
| | - Jose A. López-Guerrero
- Molecular Biology Department, Fundación Instituto Valenciano de Oncologia, Valencia, Spain
| | - David S. Moura
- Health Research Institute Fundacion Jimenez Diaz, Universidad Autonoma de Madrid, Madrid, Spain
| | - Javier Martin-Broto
- Health Research Institute Fundacion Jimenez Diaz, Universidad Autonoma de Madrid, Reyes Católicos 2, Madrid 28040, Spain
- University Hospital General de Villalba, Madrid, Spain
- Medical Oncology Department, Fundación Jimenez Diaz University Hospital, Madrid, Spain
| |
Collapse
|
5
|
Gkouvi A, Tsiogkas SG, Bogdanos DP, Gika H, Goulis DG, Grammatikopoulou MG. Proteomics in Patients with Fibromyalgia Syndrome: A Systematic Review of Observational Studies. Curr Pain Headache Rep 2024; 28:565-586. [PMID: 38652420 PMCID: PMC11271354 DOI: 10.1007/s11916-024-01244-4] [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] [Accepted: 03/16/2024] [Indexed: 04/25/2024]
Abstract
PURPOSE OF REVIEW Fibromyalgia syndrome (FMS) is a disease of unknown pathophysiology, with the diagnosis being based on a set of clinical criteria. Proteomic analysis can provide significant biological information for the pathophysiology of the disease but may also reveal biomarkers for diagnosis or therapeutic targets. The present systematic review aims to synthesize the evidence regarding the proteome of adult patients with FMS using data from observational studies. RECENT FINDINGS An extensive literature search was conducted in MEDLINE/PubMed, CENTRAL, and clinicaltrials.gov from inception until November 2022. The study protocol was published in OSF. Two independent reviewers evaluated the studies and extracted data. The quality of studies was assessed using the modified Newcastle-Ottawa scale adjusted for proteomic research. Ten studies fulfilled the protocol criteria, identifying 3328 proteins, 145 of which were differentially expressed among patients with FMS against controls. The proteins were identified in plasma, serum, cerebrospinal fluid, and saliva samples. The control groups included healthy individuals and patients with pain (inflammatory and non-inflammatory). The most important proteins identified involved transferrin, α-, β-, and γ-fibrinogen chains, profilin-1, transaldolase, PGAM1, apolipoprotein-C3, complement C4A and C1QC, immunoglobin parts, and acute phase reactants. Weak correlations were observed between proteins and pain sensation, or quality of life scales, apart from the association of transferrin and a2-macroglobulin with moderate-to-severe pain sensation. The quality of included studies was moderate-to-good. FMS appears to be related to protein dysregulation in the complement and coagulation cascades and the metabolism of iron. Several proteins may be dysregulated due to the excessive oxidative stress response.
Collapse
Affiliation(s)
- Arriana Gkouvi
- Unit of Immunonutrition and Clinical Nutrition, Department of Rheumatology and Clinical Immunology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Biopolis, Larissa, Greece
| | - Sotirios G Tsiogkas
- Unit of Immunonutrition and Clinical Nutrition, Department of Rheumatology and Clinical Immunology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Biopolis, Larissa, Greece
| | - Dimitrios P Bogdanos
- Unit of Immunonutrition and Clinical Nutrition, Department of Rheumatology and Clinical Immunology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Biopolis, Larissa, Greece.
| | - Helen Gika
- Center for Interdisciplinary Research and Innovation (CIRI-AUTH), Biomic_AUTh, Balkan Center Thermi B1.4, GR-57001, Thessaloniki, Greece
- Laboratory of Forensic Medicine and Toxicology, School of Medicine, Aristotle University of Thessaloniki, GR-54124, Thessaloniki, Greece
| | - Dimitrios G Goulis
- Unit of Reproductive Endocrinology, 1st Department of Obstetrics and Gynecology, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Maria G Grammatikopoulou
- Unit of Immunonutrition and Clinical Nutrition, Department of Rheumatology and Clinical Immunology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Biopolis, Larissa, Greece
- Unit of Reproductive Endocrinology, 1st Department of Obstetrics and Gynecology, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
6
|
Zhang R, Yao Y, Gao H, Hu X. Mechanisms of angiogenesis in tumour. Front Oncol 2024; 14:1359069. [PMID: 38590656 PMCID: PMC10999665 DOI: 10.3389/fonc.2024.1359069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 03/11/2024] [Indexed: 04/10/2024] Open
Abstract
Angiogenesis is essential for tumour growth and metastasis. Antiangiogenic factor-targeting drugs have been approved as first line agents in a variety of oncology treatments. Clinical drugs frequently target the VEGF signalling pathway during sprouting angiogenesis. Accumulating evidence suggests that tumours can evade antiangiogenic therapy through other angiogenesis mechanisms in addition to the vascular sprouting mechanism involving endothelial cells. These mechanisms include (1) sprouting angiogenesis, (2) vasculogenic mimicry, (3) vessel intussusception, (4) vascular co-option, (5) cancer stem cell-derived angiogenesis, and (6) bone marrow-derived angiogenesis. Other non-sprouting angiogenic mechanisms are not entirely dependent on the VEGF signalling pathway. In clinical practice, the conversion of vascular mechanisms is closely related to the enhancement of tumour drug resistance, which often leads to clinical treatment failure. This article summarizes recent studies on six processes of tumour angiogenesis and provides suggestions for developing more effective techniques to improve the efficacy of antiangiogenic treatment.
Collapse
Affiliation(s)
| | | | | | - Xin Hu
- China–Japan Union Hospital of Jilin University, Jilin University, Changchun, China
| |
Collapse
|
7
|
Kumar A, Nader MA, Deep G. Emergence of Extracellular Vesicles as "Liquid Biopsy" for Neurological Disorders: Boom or Bust. Pharmacol Rev 2024; 76:199-227. [PMID: 38351075 PMCID: PMC10877757 DOI: 10.1124/pharmrev.122.000788] [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: 11/23/2022] [Revised: 11/11/2023] [Accepted: 11/27/2023] [Indexed: 02/16/2024] Open
Abstract
Extracellular vesicles (EVs) have emerged as an attractive liquid biopsy approach in the diagnosis and prognosis of multiple diseases and disorders. The feasibility of enriching specific subpopulations of EVs from biofluids based on their unique surface markers has opened novel opportunities to gain molecular insight from various tissues and organs, including the brain. Over the past decade, EVs in bodily fluids have been extensively studied for biomarkers associated with various neurological disorders, such as Alzheimer's disease, Parkinson's disease, schizophrenia, bipolar disorder, major depressive disorders, substance use disorders, human immunodeficiency virus-associated neurocognitive disorder, and cancer/treatment-induced neurodegeneration. These studies have focused on the isolation and cargo characterization of either total EVs or brain cells, such as neuron-, astrocyte-, microglia-, oligodendrocyte-, pericyte-, and endothelial-derived EVs from biofluids to achieve early diagnosis and molecular characterization and to predict the treatment and intervention outcomes. The findings of these studies have demonstrated that EVs could serve as a repetitive and less invasive source of valuable molecular information for these neurological disorders, supplementing existing costly neuroimaging techniques and relatively invasive measures, like lumbar puncture. However, the initial excitement surrounding blood-based biomarkers for brain-related diseases has been tempered by challenges, such as lack of central nervous system specificity in EV markers, lengthy protocols, and the absence of standardized procedures for biological sample collection, EV isolation, and characterization. Nevertheless, with rapid advancements in the EV field, supported by improved isolation methods and sensitive assays for cargo characterization, brain cell-derived EVs continue to offer unparallel opportunities with significant translational implications for various neurological disorders. SIGNIFICANCE STATEMENT: Extracellular vesicles present a less invasive liquid biopsy approach in the diagnosis and prognosis of various neurological disorders. Characterizing these vesicles in biofluids holds the potential to yield valuable molecular information, thereby significantly impacting the development of novel biomarkers for various neurological disorders. This paper has reviewed the methodology employed to isolate extracellular vesicles derived from various brain cells in biofluids, their utility in enhancing the molecular understanding of neurodegeneration, and the potential challenges in this research field.
Collapse
Affiliation(s)
- Ashish Kumar
- Departments of Cancer Biology (A.K., G.D.), Physiology and Pharmacology (M.A.N.), Radiology (M.A.N.), and Center for Addiction Research (M.A.N., G.D.), Wake Forest University School of Medicine, Winston-Salem, North Carolina; Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, North Carolina (G.D.); and Sticht Center for Healthy Aging and Alzheimer's Prevention, Wake Forest School of Medicine, Winston-Salem, North Carolina (G.D.)
| | - Michael A Nader
- Departments of Cancer Biology (A.K., G.D.), Physiology and Pharmacology (M.A.N.), Radiology (M.A.N.), and Center for Addiction Research (M.A.N., G.D.), Wake Forest University School of Medicine, Winston-Salem, North Carolina; Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, North Carolina (G.D.); and Sticht Center for Healthy Aging and Alzheimer's Prevention, Wake Forest School of Medicine, Winston-Salem, North Carolina (G.D.)
| | - Gagan Deep
- Departments of Cancer Biology (A.K., G.D.), Physiology and Pharmacology (M.A.N.), Radiology (M.A.N.), and Center for Addiction Research (M.A.N., G.D.), Wake Forest University School of Medicine, Winston-Salem, North Carolina; Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, North Carolina (G.D.); and Sticht Center for Healthy Aging and Alzheimer's Prevention, Wake Forest School of Medicine, Winston-Salem, North Carolina (G.D.)
| |
Collapse
|
8
|
Sohn MY, Kim JW, Kang G, Woo WS, Kim KH, Son HJ, Park JW, Kim DH, Park CI. First report of L1 cell adhesion molecule (L1CAM) in flat fish (Starry flounder): Molecular features and expression analysis. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2023; 149:105054. [PMID: 37690612 DOI: 10.1016/j.dci.2023.105054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 09/07/2023] [Accepted: 09/07/2023] [Indexed: 09/12/2023]
Abstract
In this study, the starry flounder L1 cell adhesion molecule (L1CAM) sequence was obtained using next-generation sequencing, and the integrity of the sequence was verified by cloning and sequencing. First, the amino acid sequence was predicted using the cDNA sequence, and the gene was then identified through multiple sequence alignment analysis with related sequences and phylogenetic analysis. Thus, homogeneity was confirmed. The expression level of PsL1CAM (Platichthys stellatus L1CAM) mRNA in healthy starry flounder was detected in all tissues used in the experiment, and tissue- and gene-specific expression levels were confirmed. In addition, as a result of mRNA expression analysis after artificial infection with viral hemorrhagic septicemia virus (VHSV) and Streptococcus parauberis PH0710, significant expression changes and characteristics were confirmed following infection with VHSV and S. parauberis PH0710. After artificial infection with VHSV, the expression level of PsL1CAM mRNA was significantly upregulated in almost all major tissues of the starry flounder, whereas it was significantly downregulated in mucosal-associated lymphoid tissues, such as the gills and intestine. Infection with S. parauberis PH0710 significantly upregulated the expression of PsL1CAM mRNA in almost all major tissues of the starry flounder, whereas it was significantly downregulated in the heart after infection. Our results indicate that PsL1CAM may be involved in the host immune response to starry flounders.
Collapse
Affiliation(s)
- Min-Young Sohn
- Department of Marine Biology & Aquaculture, College of Marine Science, Gyeongsang National University, 455, Tongyeong, 650-160, Republic of Korea
| | - Ju-Won Kim
- Biotechnology Research Division, National Institute of Fisheries Science, Busan, 46083, Republic of Korea
| | - Gyoungsik Kang
- Department of Marine Biology & Aquaculture, College of Marine Science, Gyeongsang National University, 455, Tongyeong, 650-160, Republic of Korea
| | - Won-Sik Woo
- Department of Marine Biology & Aquaculture, College of Marine Science, Gyeongsang National University, 455, Tongyeong, 650-160, Republic of Korea
| | - Kyung-Ho Kim
- Department of Marine Biology & Aquaculture, College of Marine Science, Gyeongsang National University, 455, Tongyeong, 650-160, Republic of Korea
| | - Ha-Jeong Son
- Department of Marine Biology & Aquaculture, College of Marine Science, Gyeongsang National University, 455, Tongyeong, 650-160, Republic of Korea
| | - Jong-Won Park
- Fish Breeding Research Center, Fisheries Seed Breeding Institute, National Institute of Fisheries Science, 81-9 Geojenamseo-ro, Nambu-myeon, Geoje, 53334, Republic of Korea
| | - Do-Hyung Kim
- Department of Aquatic Life Medicine, College of Fisheries Science, Pukyong National University, 45, Yongso-ro, Nam-Gu., Busan, Republic of Korea.
| | - Chan-Il Park
- Department of Marine Biology & Aquaculture, College of Marine Science, Gyeongsang National University, 455, Tongyeong, 650-160, Republic of Korea.
| |
Collapse
|
9
|
He T, Yao Q, Xu B, Yang M, Jiang J, Xiang Q, Xiao L, Liu S, Wang H, Zhang X. A novel splicing variation in L1CAM is responsible for recurrent fetal hydrocephalus. Mol Genet Genomic Med 2023; 11:e2253. [PMID: 37489051 PMCID: PMC10655515 DOI: 10.1002/mgg3.2253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 02/20/2023] [Accepted: 07/13/2023] [Indexed: 07/26/2023] Open
Abstract
BACKGROUND The L1 cell adhesion molecule (L1CAM, OMIM 308840) gene is primarily expressed in the nervous system and encodes the L1 adhesion molecule protein. Variations in L1CAM cause a wide spectrum of X-linked neurological disorders summarized as the L1 syndrome. METHODS We report a 29-year-old pregnant woman who experienced multiple adverse pregnancy outcomes due to recurrent fetal hydrocephalus with an X-linked recessive inheritance. Genomic DNA was extracted from the third aborted male fetus and analyzed via trio whole-exome sequencing (WES). Total RNA was isolated from the pregnant woman to assess splicing variation at the mRNA level, and amniotic fluid was extracted from the woman for prenatal diagnosis on her fourth fetus. RESULTS All four male fetuses were affected by severe hydrocephalus. We identified a maternally derived hemizygous splicing variation NM_000425.5:[c.3046 + 1G > A]; NP_000416.1 p.(Gly1016AspfsTer6) (chrX:153130275) in Intron 22 of the L1CAM. This variation disrupts the donor splice site and causes the retention of Intron 22, which results in frame shift and a premature termination codon at position 1021 with the ability to produce a truncated protein without the fifth fibronectin-repeat III, transmembrane, and cytoplasmic domains or to induce the degradation of mRNAs by nonsense-mediated mRNA decay. The same hemizygous variant was also detected in the amniocytes. CONCLUSION This report enhances our knowledge of genetic and phenotypic characteristics of X-linked fetal hydrocephalus, providing a new genetic basis for prenatal diagnosis and pre-implantation prenatal diagnosis.
Collapse
Affiliation(s)
- Tiantian He
- Department of Medical Genetics & Prenatal Diagnosis CenterWest China Second University Hospital, Sichuan UniversityChengduChina
- Department of Obstetrics and GynecologyWest China Second University Hospital, Sichuan UniversityChengduChina
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of EducationChengduChina
| | - Qiang Yao
- Department of Obstetrics and GynecologyWest China Second University Hospital, Sichuan UniversityChengduChina
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of EducationChengduChina
| | - Bocheng Xu
- Department of Medical Genetics & Prenatal Diagnosis CenterWest China Second University Hospital, Sichuan UniversityChengduChina
- Department of Obstetrics and GynecologyWest China Second University Hospital, Sichuan UniversityChengduChina
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of EducationChengduChina
| | - Mei Yang
- Department of Medical Genetics & Prenatal Diagnosis CenterWest China Second University Hospital, Sichuan UniversityChengduChina
- Department of Obstetrics and GynecologyWest China Second University Hospital, Sichuan UniversityChengduChina
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of EducationChengduChina
| | - Jieni Jiang
- Department of Medical Genetics & Prenatal Diagnosis CenterWest China Second University Hospital, Sichuan UniversityChengduChina
- Department of Obstetrics and GynecologyWest China Second University Hospital, Sichuan UniversityChengduChina
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of EducationChengduChina
| | - Qingqing Xiang
- Department of Medical Genetics & Prenatal Diagnosis CenterWest China Second University Hospital, Sichuan UniversityChengduChina
- Department of Obstetrics and GynecologyWest China Second University Hospital, Sichuan UniversityChengduChina
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of EducationChengduChina
| | - Like Xiao
- Department of Medical Genetics & Prenatal Diagnosis CenterWest China Second University Hospital, Sichuan UniversityChengduChina
- Department of Obstetrics and GynecologyWest China Second University Hospital, Sichuan UniversityChengduChina
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of EducationChengduChina
| | - Shanling Liu
- Department of Medical Genetics & Prenatal Diagnosis CenterWest China Second University Hospital, Sichuan UniversityChengduChina
- Department of Obstetrics and GynecologyWest China Second University Hospital, Sichuan UniversityChengduChina
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of EducationChengduChina
| | - He Wang
- Department of Medical Genetics & Prenatal Diagnosis CenterWest China Second University Hospital, Sichuan UniversityChengduChina
- Department of Obstetrics and GynecologyWest China Second University Hospital, Sichuan UniversityChengduChina
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of EducationChengduChina
| | - Xuemei Zhang
- Department of Medical Genetics & Prenatal Diagnosis CenterWest China Second University Hospital, Sichuan UniversityChengduChina
- Department of Obstetrics and GynecologyWest China Second University Hospital, Sichuan UniversityChengduChina
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of EducationChengduChina
| |
Collapse
|
10
|
Dos Santos MV, Holth A, Lindemann K, Staff AC, Davidson B. Clinical significance of L1CAM expression in metastatic tubo-ovarian high-grade serous carcinoma. Gynecol Oncol 2023; 176:76-81. [PMID: 37478615 DOI: 10.1016/j.ygyno.2023.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 06/30/2023] [Accepted: 07/04/2023] [Indexed: 07/23/2023]
Abstract
OBJECTIVE To analyze the expression and prognostic role of L1CAM in tubo-ovarian high-grade serous carcinoma (HGSC). METHODS L1CAM protein expression by immunohistochemistry was analyzed in 644 HGSC (413 effusions, 231 surgical specimens). Expression was analyzed for association with clinicopathologic parameters and survival. RESULTS L1CAM protein expression was found in 401/413 (97%) effusions and 209/231 (90%) surgical specimens, with significantly higher staining extent in effusions (p < 0.001). L1CAM protein expression in effusions was unrelated to clinicopathologic parameters (p > 0.05). In surgical specimens, higher L1CAM expression was significantly related to primary (intrinsic) chemoresistance (p = 0.017). High (>25%) L1CAM expression in HGSC effusions (p = 0.02), older patient age (p = 0.013), FIGO stage IV disease (p < 0.001) and larger residual disease volume (p = 0.001) were significantly associated with shorter overall survival (OS) in univariate analysis. In Cox multivariate analysis, only FIGO stage (p = 0.001) and residual disease volume (p = 0.003) were independent prognosticators of OS. L1CAM expression in effusions was unrelated to progression-free survival (PFS). There was no association between L1CAM expression in surgical specimens and survival. CONCLUSION L1CAM is overexpressed in HGSC effusions compared to surgical specimens. Its overexpression in effusions is significantly associated with shorter OS, but not independently of established prognostic factors such as FIGO stage and residual disease volume.
Collapse
Affiliation(s)
| | - Arild Holth
- Department of Pathology, Oslo University Hospital, Norwegian Radium Hospital, N-0310 Oslo, Norway
| | - Kristina Lindemann
- University of Oslo, Faculty of Medicine, Institute of Clinical Medicine, N-0316 Oslo, Norway; Department of Gynecologic Oncology, Oslo University Hospital, Norwegian Radium Hospital, N-0310 Oslo, Norway
| | - Anne Cathrine Staff
- University of Oslo, Faculty of Medicine, Institute of Clinical Medicine, N-0316 Oslo, Norway; Department of Obstetrics and Gynecology, Oslo University Hospital, Ullevål Hospital, N-0450 Oslo, Norway
| | - Ben Davidson
- Department of Pathology, Oslo University Hospital, Norwegian Radium Hospital, N-0310 Oslo, Norway; University of Oslo, Faculty of Medicine, Institute of Clinical Medicine, N-0316 Oslo, Norway.
| |
Collapse
|
11
|
Yang H, Gu X, Chen H, Zeng Q, Mao Z, Ge Y. Omics techniques reveal the toxicity mechanisms of three antiepileptic drugs to juvenile zebrafish (Danio rerio) brain and liver. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2023; 262:106668. [PMID: 37659109 DOI: 10.1016/j.aquatox.2023.106668] [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: 07/04/2023] [Revised: 08/18/2023] [Accepted: 08/21/2023] [Indexed: 09/04/2023]
Abstract
Epilepsy, a neurological disorder, is characterized by seizures that are an appearance of excessive brain activity and is symptomatically treated with antiepileptic drugs (AEDs). Oxcarbazepine (OCBZ), lamotrigine (LTG), and carbamazepine (CBZ) are widely used AEDs in clinics and are very often detected in aquatic environments. However, neither the sub-lethal effects nor the specific mechanisms of these AEDs' action on the fish are well understood. In this study, juvenile zebrafish were exposed to a sub-lethal concentration (100 μg/L) of OCBZ, LTG, and CBZ for 28 d, after which indicators of oxidative stress (i.e. superoxide dismutase (SOD) activity, catalase (CAT) activity, and malondialdehyde (MDA) level) and neurotoxicity (i.e. acetylcholinesterase (AChE) activity, γ-aminobutyric acid (GABA) level, and glutamic acid (Glu) level) were measured. Brain SOD activity was significantly increased by three AEDs, while brain CAT activity was significantly inhibited by LTG and CBZ. Liver SOD activity was significantly enhanced by CBZ, and liver CAT activity was significantly induced by OCBZ and LTG. Liver MDA level was significantly increased by three AEDs. Brain AChE activity was significantly increased by LTG and CBZ, and brain GABA level was significantly enhanced by three AEDs. However, there were no significant alterations in the levels of MDA and Glu in zebrafish brain. To ascertain mechanisms of AEDs-induced toxicity, brain transcriptomics and liver metabolomics were conducted in zebrafish. The brain transcriptomics results showed that lots of differentially expressed genes (DEGs) were enriched in the sensory system, the immune system, the digestive system, the metabolic processes, and others in three AEDs treated groups. The metabolomics data indicated dysregulation of glycerophospholipid signaling and lipid homeostasis in zebrafish liver after three AEDs exposure. The overall results of this study improve understanding of the sub-lethal effects and potential molecular mechanisms of action of AEDs in fish.
Collapse
Affiliation(s)
- Huiting Yang
- State Key Laboratory of Lake Science and Environment, Nanjing Institute of Geography and Limnology, Chinese Academy of Sciences, Nanjing 210008, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaohong Gu
- State Key Laboratory of Lake Science and Environment, Nanjing Institute of Geography and Limnology, Chinese Academy of Sciences, Nanjing 210008, China.
| | - Huihui Chen
- State Key Laboratory of Lake Science and Environment, Nanjing Institute of Geography and Limnology, Chinese Academy of Sciences, Nanjing 210008, China
| | - Qingfei Zeng
- State Key Laboratory of Lake Science and Environment, Nanjing Institute of Geography and Limnology, Chinese Academy of Sciences, Nanjing 210008, China
| | - Zhigang Mao
- State Key Laboratory of Lake Science and Environment, Nanjing Institute of Geography and Limnology, Chinese Academy of Sciences, Nanjing 210008, China
| | - You Ge
- State Key Laboratory of Lake Science and Environment, Nanjing Institute of Geography and Limnology, Chinese Academy of Sciences, Nanjing 210008, China; University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
12
|
Gajendran N, Rajasekaran S, Witt SN. Knocking out alpha-synuclein in melanoma cells downregulates L1CAM and decreases motility. Sci Rep 2023; 13:9243. [PMID: 37286800 DOI: 10.1038/s41598-023-36451-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 06/03/2023] [Indexed: 06/09/2023] Open
Abstract
The Parkinson's disease (PD) associated protein, alpha-synuclein (α-syn/SNCA), is highly expressed in aggressive melanomas. The goal of this study was to reveal possible mechanism(s) of α-syn involvement in melanoma pathogenesis. Herein, we asked whether α-syn modulates the expression of the pro-oncogenic adhesion molecules L1CAM and N-cadherin. We used two human melanoma cell lines (SK-MEL-28, SK-MEL-29), SNCA-knockout (KO) clones, and two human SH-SY5Y neuroblastoma cell lines. In the melanoma lines, loss of α-syn expression resulted in significant decreases in the expression of L1CAM and N-cadherin and concomitant significant decreases in motility. On average, there was a 75% reduction in motility in the four SNCA-KOs tested compared to control cells. Strikingly, comparing neuroblastoma SH-SY5Y cells that have no detectable α-syn to SH-SY5Y cells that stably express α-syn (SH/+αS), we found that expressing α-syn increased L1CAM and single-cell motility by 54% and 597%, respectively. The reduction in L1CAM level in SNCA-KO clones was not due to a transcriptional effect, rather we found that L1CAM is more efficiently degraded in the lysosome in SNCA-KO clones than in control cells. We propose that α-syn is pro-survival to melanoma (and possibly neuroblastoma) because it promotes the intracellular trafficking of L1CAM to the plasma membrane.
Collapse
Affiliation(s)
- Nithya Gajendran
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Santhanasabapathy Rajasekaran
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Stephan N Witt
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, USA.
- Feist-Weiller Cancer Center, Louisiana State University Health Shreveport, Shreveport, USA.
| |
Collapse
|
13
|
Li L, Wang X, Hu K, Liu X, Qiu L, Bai C, Cui Y, Wang B, Wang Z, Wang H, Cheng R, Hua J, Hai L, Wang M, Liu M, Song Z, Xiao C, Li B. ZNF133 is a potent suppressor in breast carcinogenesis through dampening L1CAM, a driver for tumor progression. Oncogene 2023:10.1038/s41388-023-02731-5. [PMID: 37221223 DOI: 10.1038/s41388-023-02731-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 05/04/2023] [Accepted: 05/12/2023] [Indexed: 05/25/2023]
Abstract
Due to the complexity and heterogeneity of breast cancer, the therapeutic effects of breast cancer treatment vary between subtypes. Breast cancer subtypes are classified based on the presence of molecular markers for estrogen or progesterone receptors and human epidermal growth factor 2. Thus, novel, comprehensive, and precise molecular indicators in breast carcinogenesis are urgently needed. Here, we report that ZNF133, a zinc-finger protein, is negatively associated with poor survival and advanced pathological staging of breast carcinomas. Moreover, ZNF133 is a transcription repressor physically associated with the KAP1 complex. It transcriptionally represses a cohort of genes, including L1CAM, that are critically involved in cell proliferation and motility. We also demonstrate that the ZNF133/KAP1 complex inhibits the proliferation and invasion of breast cancer cells in vitro and suppresses breast cancer growth and metastasis in vivo by dampening the transcription of L1CAM. Taken together, the findings of our study confirm the value of ZNF133 and L1CAM levels in the diagnosis and prognosis of breast cancer, contribute to a deeper understanding of the regulation mechanism of ZNF133 for the first time, and provide a new therapeutic strategy and precise intervention target for breast cancer.
Collapse
Affiliation(s)
- Lifang Li
- Department of Cancer Cell Biology, Tianjin's Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, PR China.
| | - Xuefei Wang
- Department of Cancer Cell Biology, Tianjin's Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, PR China
| | - Kai Hu
- Department of Pathology, School of Medicine, Nankai University, Tianjin, 300071, PR China
| | - Xinhua Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou, 311121, PR China
| | - Li Qiu
- Department of Cancer Cell Biology, Tianjin's Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, PR China
| | - Changsen Bai
- Department of Clinical Laboratory, Tianjin's Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, PR China
| | - Yanfen Cui
- Public Laboratory, Tianjin's Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, PR China
| | - Biyun Wang
- Laboratory Animal Center, Tianjin's Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, PR China
| | - Zhaosong Wang
- Laboratory Animal Center, Tianjin's Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, PR China
| | - Hailong Wang
- Department of Cancer Cell Biology, Tianjin's Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, PR China
| | - Runfen Cheng
- Department of Cancer Cell Biology, Tianjin's Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, PR China
| | - Jialei Hua
- Department of Cancer Cell Biology, Tianjin's Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, PR China
| | - Linyue Hai
- Department of Cancer Cell Biology, Tianjin's Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, PR China
| | - Mengdie Wang
- Department of Cancer Cell Biology, Tianjin's Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, PR China
| | - Miao Liu
- Department of Cancer Cell Biology, Tianjin's Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, PR China
| | - Zian Song
- Department of Cancer Cell Biology, Tianjin's Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, PR China
| | - Chunhua Xiao
- First Surgical Department of Breast Cancer, Tianjin's Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, PR China.
| | - Binghui Li
- Department of Cancer Cell Biology, Tianjin's Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, PR China.
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, PR China.
| |
Collapse
|
14
|
Dutta S, Hornung S, Taha HB, Bitan G. Biomarkers for parkinsonian disorders in CNS-originating EVs: promise and challenges. Acta Neuropathol 2023; 145:515-540. [PMID: 37012443 PMCID: PMC10071251 DOI: 10.1007/s00401-023-02557-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/27/2023] [Accepted: 03/07/2023] [Indexed: 04/05/2023]
Abstract
Extracellular vesicles (EVs), including exosomes, microvesicles, and oncosomes, are nano-sized particles enclosed by a lipid bilayer. EVs are released by virtually all eukaryotic cells and have been shown to contribute to intercellular communication by transporting proteins, lipids, and nucleic acids. In the context of neurodegenerative diseases, EVs may carry toxic, misfolded forms of amyloidogenic proteins and facilitate their spread to recipient cells in the central nervous system (CNS). CNS-originating EVs can cross the blood-brain barrier into the bloodstream and may be found in other body fluids, including saliva, tears, and urine. EVs originating in the CNS represent an attractive source of biomarkers for neurodegenerative diseases, because they contain cell- and cell state-specific biological materials. In recent years, multiple papers have reported the use of this strategy for identification and quantitation of biomarkers for neurodegenerative diseases, including Parkinson's disease and atypical parkinsonian disorders. However, certain technical issues have yet to be standardized, such as the best surface markers for isolation of cell type-specific EVs and validating the cellular origin of the EVs. Here, we review recent research using CNS-originating EVs for biomarker studies, primarily in parkinsonian disorders, highlight technical challenges, and propose strategies for overcoming them.
Collapse
Affiliation(s)
- Suman Dutta
- International Institute of Innovation and Technology, New Town, Kolkata, India
| | - Simon Hornung
- Division of Peptide Biochemistry, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Hash Brown Taha
- Department of Integrative Biology and Physiology, University of California Los Angeles, Los Angeles, CA, USA
- Department of Neurology, David Geffen School of Medicine at UCLA, University of California Los Angeles, 635 Charles E. Young Drive South/Gordon 451, Los Angeles, CA, 90095, USA
| | - Gal Bitan
- Department of Neurology, David Geffen School of Medicine at UCLA, University of California Los Angeles, 635 Charles E. Young Drive South/Gordon 451, Los Angeles, CA, 90095, USA.
- Brain Research Institute, University of California Los Angeles, Los Angeles, CA, USA.
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
15
|
Specificity of viscumin revised. As probed with a printed glycan array. Biochimie 2022; 202:94-102. [PMID: 35988841 DOI: 10.1016/j.biochi.2022.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/11/2022] [Accepted: 08/13/2022] [Indexed: 01/01/2023]
Abstract
Viscumin, a lectin used in anti-cancer therapy, was originally considered as βGal recognizing protein; later, an ability to bind 6'-sialyl N-acetyllactosamine (6'SLN) terminated gangliosides was found. Here we probed viscumin with a printed glycan array (PGA) containing a large number of mammalian sulfated glycans, and found a strong binding to glycans with 6-O-SuGal moiety as lactose, N-acetyllactosamine (LN), di-N-acetyllactosamine (LacdiNAc), and even 6-O-SuGalNAcα (but not SiaTn). Also, the ability to bind some of αGal terminated glycans, including Galα1-3Galβ1-4GlcNAc, was observed. Unexpectedly, only weak interaction was detected with parent neutral β-galactosides including LN-LN-LN and branched (LN)2LN oligolactosamines; in the light of these data, one should not confidently classify viscumin as a β-galactoside-binding lectin. Carrying out PGA in the presence of neutral or sulfated/sialylated glycan, together with sequential elution from lactose-sepharose and consideration of the protein structure, lead to the conclusion that two glycan-binding sites of viscumin have different specificities, one of which prefers charged sulfated and sialylated moieties.
Collapse
|
16
|
Uncovering Novel Features of the Pc Locus in Horn Development from Gene-Edited Holstein Cattle by RNA-Sequencing Analysis. Int J Mol Sci 2022; 23:ijms232012060. [PMID: 36292916 PMCID: PMC9603690 DOI: 10.3390/ijms232012060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 10/01/2022] [Accepted: 10/04/2022] [Indexed: 11/19/2022] Open
Abstract
The Polled Celtic (Pc) mutation locus is a genetically simple single mutation that is the best choice for breeding polled cattle using gene editing. However, the mechanism of the Pc locus for regulating horn development is unclear, so we used gene editing, somatic cell nuclear transfer and embryo transfer to obtain polled Holstein fetal bovine (gestation time 90 days) with a homozygous Pc insertion (gene-edited Holstein fetal bovine, EH) and the wild-type 90 days Holstein fetal bovine (WH) as controls. The hematoxylin-eosin (HE) staining results showed that, compared to the WH, the EH horn buds had no white keratinized projections or vacuolated keratinocytes and no thick nerve bundles under the dermal tissue. Furthermore, DNA sequencing results showed that the Pc locus was homozygously inserted into the fetal bovine genome. A total of 791 differentially expressed genes were identified by transcriptome sequencing analysis. Enrichment analysis and protein interaction analysis results of differentially expressed genes showed that abundant gene changes after Pc insertion were associated with the adhesion molecule regulation, actin expression, cytoskeletal deformation and keratin expression and keratinization. It was also noted that the results contained several genes that had been reported to be associated with the development of horn traits, such as RXFP2 and TWIST1. This study identified these changes for the first time and summarized them. The results suggested that the Pc mutant locus may inhibit neural crest cell EMT generation and keratin expression, leading to failures in neural crest cell migration and keratinization of the horn bud tissue, regulating the production of the polled phenotype.
Collapse
|
17
|
Zhang B, Ma X, Huang B, Jiang Q, Loor JJ, Lv X, Zhang W, Li M, Wen J, Yin Y, Wang J, Yang W, Xu C. Transcriptomics of circulating neutrophils in dairy cows with subclinical hypocalcemia. Front Vet Sci 2022; 9:959831. [PMID: 36176696 PMCID: PMC9514324 DOI: 10.3389/fvets.2022.959831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 07/20/2022] [Indexed: 11/13/2022] Open
Abstract
Hypocalcemia is closely associated with inflammatory diseases in dairy cows. Recent research has underscored the key role of calcium in the adaptations of the innate immune system during this period. The main objective in the present study was to compare the transcriptome profiles and analyze differences in the expression of neutrophil (PMNL) immune function-related genes and calcium binding-related genes in hypocalcemic cows. At 2 days postpartum, a concentration >2.10 mmol Ca2+/L was used to classify cows as controls (CON), and a concentration <2.00 mmol Ca2+/L used to classify cows as low-calcium (LCAL) (n = 8 in each group). A routine medical examination was conducted by the attending veterinarian to ensure there were no other complications and that the blood β-hydroxybutyrate was <1.2 mmol/L. Blood was collected from the tail vein (20 mL) to isolate PMNL, and 5 cows in each group were used for RNA sequencing and statistical analysis of gene expression differences. Transcriptome RNA-seq sequencing analysis was via omicsstudio using the R package edgeR. GO and KEGG enrichment analysis were used for bioinformatics. The remaining 3 cows in each group were used for validation of RNA sequencing data via quantitative PCR, which confirmed the observed responses. Compared with CON, 158 genes in LCAL were significantly up-regulated and 296 genes were down-regulated. The downregulation of Interleukin-12 (CXCL12), Tubulin beta chain (TUBB1), L1 cell adhesion molecule (L1CAM), and Myeloperoxidase (MPO) indicated a decrease in immune function of PMNL in LCAL cows. The decreased expression of calcium-binding pathway-related genes in PMNL of LCAL cows indicated a decrease in immune function of PMNL likely related to calcium ions. For example, cartilage acid protein 1 (CRTAC1) and calcium/calmodulin-dependent kinase 4 (CAMK4) were significantly reduced in LCAL cows. The upregulation of Cyclin dependent kinase inhibitor 1A (CDKN1A), Perforin 1 (PRF1), and Homeodomain interacting protein kinase 3 (HIPK3) indicated that LCAL led to greater cell apoptosis and senescence. Overall, the analyses indicated that the reduction in PMNL immune function during hypocalcemia is associated with downregulation of intracellular Ca2+ related genes and upregulation of genes controlling apoptosis and senescence. Together, these alterations contribute to an immunosuppressive state during the transition period.
Collapse
Affiliation(s)
- Bingbing Zhang
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Xinru Ma
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Baoyin Huang
- Animal Husbandry and Veterinary Branch of Heilongjiang Academy of Agricultural Sciences, Qiqihaer, China
| | - Qianming Jiang
- Mammalian NutriPhysioGenomics, Division of Nutritional Sciences, Department of Animal Sciences, University of Illinois, Urbana, IL, United States
| | - Juan J. Loor
- Mammalian NutriPhysioGenomics, Division of Nutritional Sciences, Department of Animal Sciences, University of Illinois, Urbana, IL, United States
| | - Xinquan Lv
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Wei Zhang
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Ming Li
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Jianan Wen
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Yufeng Yin
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Jingjing Wang
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Wei Yang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Chuang Xu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
- College of Veterinary Medicine, China Agricultural University, Beijing, China
- *Correspondence: Chuang Xu
| |
Collapse
|
18
|
Novosad VO, Polikanova IS, Tonevitsky EA, Maltseva DV. Expression of CD44 Isoforms in Tumor Samples and Cell Lines of Human Colorectal Cancer. Bull Exp Biol Med 2022; 173:155-159. [PMID: 35618971 DOI: 10.1007/s10517-022-05512-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Indexed: 10/18/2022]
Abstract
Detection of colorectal cancer biomarkers (CRC) remains an urgent task for the diagnosis and prediction of the disease course. A promising approach is the study of cancer stem cell markers. The cell surface glycoprotein CD44 is very important for CRC and its stem cells. Alternative splicing of 9 variable exons of CD44 mRNA leads to the formation of various isoforms of the protein with different roles in the progression of cancer. Studies of the functions of CD44 isoforms require adequate models considering the distribution of CD44 isoforms in real tumor samples. In the present study, the expression profile of CD44 isoforms in CRC was assessed based on the publicly available mRNA sequencing data of patient tumors from the TCGA-COAD database. It was shown that normal tissues predominantly expressed isoforms 3 and 4 at nearly equal levels, whereas tumors mainly expressed isoforms 2, 3, and 4; isoform 3 was expressed at the highest level. Further, the most relevant cell lines for studying the role of CD44 in CRC were identified based on the analysis of mRNA sequencing data of 55 CRC cell lines form CCLE database.
Collapse
Affiliation(s)
- V O Novosad
- Faculty of Biology and Biotechnologies, National Research University Higher School of Economics (HSE University), Moscow, Russia.
| | - I S Polikanova
- Faculty of Biology and Biotechnologies, National Research University Higher School of Economics (HSE University), Moscow, Russia
| | - E A Tonevitsky
- Faculty of Biology and Biotechnologies, National Research University Higher School of Economics (HSE University), Moscow, Russia
| | - D V Maltseva
- Faculty of Biology and Biotechnologies, National Research University Higher School of Economics (HSE University), Moscow, Russia
| |
Collapse
|
19
|
Sologova SS, Zavadskiy SP, Mokhosoev IM, Moldogazieva NT. Short Linear Motifs Orchestrate Functioning of Human Proteins during Embryonic Development, Redox Regulation, and Cancer. Metabolites 2022; 12:metabo12050464. [PMID: 35629968 PMCID: PMC9144484 DOI: 10.3390/metabo12050464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/18/2022] [Accepted: 05/19/2022] [Indexed: 11/16/2022] Open
Abstract
Short linear motifs (SLiMs) are evolutionarily conserved functional modules of proteins that represent amino acid stretches composed of 3 to 10 residues. The biological activities of two short peptide segments of human alpha-fetoprotein (AFP), a major embryo-specific and cancer-related protein, have been confirmed experimentally. This is a heptapeptide segment LDSYQCT in domain I designated as AFP14–20 and a nonapeptide segment EMTPVNPGV in domain III designated as GIP-9. In our work, we searched the UniprotKB database for human proteins that contain SLiMs with sequence similarity to the both segments of human AFP and undertook gene ontology (GO)-based functional categorization of retrieved proteins. Gene set enrichment analysis included GO terms for biological process, molecular function, metabolic pathway, KEGG pathway, and protein–protein interaction (PPI) categories. We identified the SLiMs of interest in a variety of non-homologous proteins involved in multiple cellular processes underlying embryonic development, cancer progression, and, unexpectedly, the regulation of redox homeostasis. These included transcription factors, cell adhesion proteins, ubiquitin-activating and conjugating enzymes, cell signaling proteins, and oxidoreductase enzymes. They function by regulating cell proliferation and differentiation, cell cycle, DNA replication/repair/recombination, metabolism, immune/inflammatory response, and apoptosis. In addition to the retrieved genes, new interacting genes were identified. Our data support the hypothesis that conserved SLiMs are incorporated into non-homologous proteins to serve as functional blocks for their orchestrated functioning.
Collapse
Affiliation(s)
- Susanna S. Sologova
- Nelyubin Institute of Pharmacy, Sechenov First Moscow State Medical University, (Sechenov University), 119991 Moscow, Russia; (S.S.S.); (S.P.Z.)
| | - Sergey P. Zavadskiy
- Nelyubin Institute of Pharmacy, Sechenov First Moscow State Medical University, (Sechenov University), 119991 Moscow, Russia; (S.S.S.); (S.P.Z.)
| | - Innokenty M. Mokhosoev
- Department of Biochemistry and Molecular Biology, Pirogov Russian National Research Medical University, 117997 Moscow, Russia;
| | - Nurbubu T. Moldogazieva
- Nelyubin Institute of Pharmacy, Sechenov First Moscow State Medical University, (Sechenov University), 119991 Moscow, Russia; (S.S.S.); (S.P.Z.)
- Correspondence:
| |
Collapse
|
20
|
Bi-allelic variants in neuronal cell adhesion molecule cause a neurodevelopmental disorder characterized by developmental delay, hypotonia, neuropathy/spasticity. Am J Hum Genet 2022; 109:518-532. [PMID: 35108495 DOI: 10.1016/j.ajhg.2022.01.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 01/07/2022] [Indexed: 12/20/2022] Open
Abstract
Cell adhesion molecules are membrane-bound proteins predominantly expressed in the central nervous system along principal axonal pathways with key roles in nervous system development, neural cell differentiation and migration, axonal growth and guidance, myelination, and synapse formation. Here, we describe ten affected individuals with bi-allelic variants in the neuronal cell adhesion molecule NRCAM that lead to a neurodevelopmental syndrome of varying severity; the individuals are from eight families. This syndrome is characterized by developmental delay/intellectual disability, hypotonia, peripheral neuropathy, and/or spasticity. Computational analyses of NRCAM variants, many of which cluster in the third fibronectin type III (Fn-III) domain, strongly suggest a deleterious effect on NRCAM structure and function, including possible disruption of its interactions with other proteins. These findings are corroborated by previous in vitro studies of murine Nrcam-deficient cells, revealing abnormal neurite outgrowth, synaptogenesis, and formation of nodes of Ranvier on myelinated axons. Our studies on zebrafish nrcamaΔ mutants lacking the third Fn-III domain revealed that mutant larvae displayed significantly altered swimming behavior compared to wild-type larvae (p < 0.03). Moreover, nrcamaΔ mutants displayed a trend toward increased amounts of α-tubulin fibers in the dorsal telencephalon, demonstrating an alteration in white matter tracts and projections. Taken together, our study provides evidence that NRCAM disruption causes a variable form of a neurodevelopmental disorder and broadens the knowledge on the growing role of the cell adhesion molecule family in the nervous system.
Collapse
|
21
|
Gomes DE, Witwer KW. L1CAM-associated extracellular vesicles: A systematic review of nomenclature, sources, separation, and characterization. JOURNAL OF EXTRACELLULAR BIOLOGY 2022; 1:e35. [PMID: 35492832 PMCID: PMC9045013 DOI: 10.1002/jex2.35] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 02/19/2022] [Accepted: 02/24/2022] [Indexed: 12/20/2022]
Abstract
When released into biological fluids like blood or saliva, brain extracellular vesicles (EVs) might provide a window into otherwise inaccessible tissue, contributing useful biomarkers of neurodegenerative and other central nervous system (CNS) diseases. To enrich for brain EVs in the periphery, however, cell-specific EV surface markers are needed. The protein that has been used most frequently to obtain EVs of putative neuronal origin is the transmembrane L1 cell adhesion molecule (L1CAM/CD171). In this systematic review, we examine the existing literature on L1CAM and EVs, including investigations of both neurodegenerative disease and cancer through the lens of the minimal information for studies of EVs (MISEV), specifically in the domains of nomenclature usage, EV sources, and EV separation and characterization. Although numerous studies have reported L1CAM-associated biomarker signatures that correlate with disease, interpretation of these results is complicated since L1CAM expression is not restricted to neurons and is also upregulated during cancer progression. A recent study has suggested that L1CAM epitopes are present in biofluids mostly or entirely as cleaved, soluble protein. Our findings on practices and trends in L1CAM-mediated EV separation, enrichment, and characterization yield insights that may assist with interpreting results, evaluating rigor, and suggesting avenues for further exploration.
Collapse
Affiliation(s)
- Dimitria E. Gomes
- Cornell University College of Veterinary MedicineIthacaNew YorkUSA
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Kenneth W. Witwer
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of NeurologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- The Richman Family Precision Medicine Centre of Excellence in Alzheimer's DiseaseJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| |
Collapse
|
22
|
Transcription Factors Leading to High Expression of Neuropeptide L1CAM in Brain Metastases from Lung Adenocarcinoma and Clinical Prognostic Analysis. DISEASE MARKERS 2022; 2021:8585633. [PMID: 35003395 PMCID: PMC8739529 DOI: 10.1155/2021/8585633] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 12/04/2021] [Accepted: 12/13/2021] [Indexed: 12/17/2022]
Abstract
Background There is a lack of understanding of the development of metastasis in lung adenocarcinoma (LUAD). This study is aimed at exploring the upstream regulatory transcription factors of L1 cell adhesion molecule (L1CAM) and to construct a prognostic model to predict the risk of brain metastasis in LUAD. Methods Differences in gene expression between LUAD and brain metastatic LUAD were analyzed using the Wilcoxon rank-sum test. The GRNdb (http://www.grndb.com) was used to reveal the upstream regulatory transcription factors of L1CAM in LUAD. Single-cell expression profile data (GSE131907) were obtained from the transcriptome data of 10 metastatic brain tissue samples. LUAD prognostic nomogram prediction models were constructed based on the identified significant transcription factors and L1CAM. Results Survival analysis suggested that high L1CAM expression was negatively significantly associated with overall survival, disease-specific survival, and prognosis in the progression-free interval (p < 0.05). The box plot indicates that high expression of L1CAM was associated with distant metastases in LUAD, while ROC curves suggested that high expression of L1CAM was associated with poor prognosis. FOSL2, HOXA9, IRF4, IKZF1, STAT1, FLI1, ETS1, E2F7, and ADARB1 are potential upstream transcriptional regulators of L1CAM. Single-cell data analysis revealed that the expression of L1CAM was found significantly and positively correlated with the expression of ETS1, FOSL2, and STAT1 in brain metastases. L1CAM, ETS1, FOSL2, and STAT1 were used to construct the LUAD prognostic nomogram prediction model, and the ROC curves suggest that the constructed nomogram possesses good predictive power. Conclusion By bioinformatics methods, ETS1, FOSL2, and STAT1 were identified as potential transcriptional regulators of L1CAM in this study. This will help to facilitate the early identification of patients at high risk of metastasis.
Collapse
|
23
|
Bando H, Urai S, Kanie K, Sasaki Y, Yamamoto M, Fukuoka H, Iguchi G, Camper SA. Novel genes and variants associated with congenital pituitary hormone deficiency in the era of next-generation sequencing. Front Endocrinol (Lausanne) 2022; 13:1008306. [PMID: 36237189 PMCID: PMC9551393 DOI: 10.3389/fendo.2022.1008306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 09/09/2022] [Indexed: 01/07/2023] Open
Abstract
Combined pituitary hormone deficiency (CPHD) is not a rare disorder, with a frequency of approximately 1 case per 4,000 live births. However, in most cases, a genetic diagnosis is not available. Furthermore, the diagnosis is challenging because no clear correlation exists between the pituitary hormones affected and the gene(s) responsible for the disorder. Next-generation sequencing (NGS) has recently been widely used to identify novel genes that cause (or putatively cause) CPHD. This review outlines causative genes for CPHD that have been newly reported in recent years. Moreover, novel variants of known CPHD-related genes (POU1F1 and GH1 genes) that contribute to CPHD through unique mechanisms are also discussed in this review. From a clinical perspective, variants in some of the recently identified causative genes result in extra-pituitary phenotypes. Clinical research on the related symptoms and basic research on pituitary formation may help in inferring the causative gene(s) of CPHD. Future NGS analysis of a large number of CPHD cases may reveal new genes related to pituitary development. Clarifying the causative genes of CPHD may help to understand the process of pituitary development. We hope that future innovations will lead to the identification of genes responsible for CPHD and pituitary development.
Collapse
Affiliation(s)
- Hironori Bando
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Hospital, Kobe, Japan
- *Correspondence: Hironori Bando,
| | - Shin Urai
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University School of Medicine, Kobe, Japan
| | - Keitaro Kanie
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Hospital, Kobe, Japan
| | - Yuriko Sasaki
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Hospital, Kobe, Japan
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University School of Medicine, Kobe, Japan
| | - Masaaki Yamamoto
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Hospital, Kobe, Japan
| | - Hidenori Fukuoka
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Hospital, Kobe, Japan
| | - Genzo Iguchi
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Hospital, Kobe, Japan
- Division of Biosignal Pathophysiology, Kobe University Graduate School of Medicine, Kobe, Japan
- Medical Center for Student Health, Kobe University, Kobe, Japan
| | - Sally A. Camper
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
24
|
Huang Y, Driedonks TA, Cheng L, Rajapaksha H, Routenberg DA, Nagaraj R, Redding J, Arab T, Powell BH, Pletniková O, Troncoso JC, Zheng L, Hill AF, Mahairaki V, Witwer KW. Brain Tissue-Derived Extracellular Vesicles in Alzheimer's Disease Display Altered Key Protein Levels Including Cell Type-Specific Markers. J Alzheimers Dis 2022; 90:1057-1072. [PMID: 36213994 PMCID: PMC9741741 DOI: 10.3233/jad-220322] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/02/2022] [Indexed: 11/05/2022]
Abstract
BACKGROUND Brain tissue-derived extracellular vesicles (bdEVs) play neurodegenerative and protective roles, including in Alzheimer's disease (AD). Extracellular vesicles (EVs) may also leave the brain to betray the state of the CNS in the periphery. Only a few studies have profiled the proteome of bdEVs and source brain tissue. Additionally, studies focusing on bdEV cell type-specific surface markers are rare. OBJECTIVE We aimed to reveal the pathological mechanisms inside the brain by profiling the tissue and bdEV proteomes in AD patients. In addition, to indicate targets for capturing and molecular profiling of bdEVs in the periphery, CNS cell-specific markers were profiled on the intact bdEV surface. METHODS bdEVs were separated and followed by EV counting and sizing. Brain tissue and bdEVs from age-matched AD patients and controls were then proteomically profiled. Total tau (t-tau), phosphorylated tau (p-tau), and antioxidant peroxiredoxins (PRDX) 1 and 6 were measured by immunoassay in an independent bdEV separation. Neuron, microglia, astrocyte, and endothelia markers were detected on intact EVs by multiplexed ELISA. RESULTS Overall, concentration of recovered bdEVs was not affected by AD. Proteome differences between AD and control were more pronounced for bdEVs than for brain tissue. Levels of t-tau, p-tau, PRDX1, and PRDX6 were significantly elevated in AD bdEVs compared with controls. Release of certain cell-specific bdEV markers was increased in AD. CONCLUSION Several bdEV proteins are involved in AD mechanisms and may be used for disease monitoring. The identified CNS cell markers may be useful tools for peripheral bdEV capture.
Collapse
Affiliation(s)
- Yiyao Huang
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Tom A.P. Driedonks
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lesley Cheng
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC, Australia
| | - Harinda Rajapaksha
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC, Australia
| | | | | | - Javier Redding
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Tanina Arab
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Bonita H. Powell
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Olga Pletniková
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Juan C. Troncoso
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lei Zheng
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Andrew F. Hill
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC, Australia
- Institute of Health and Sport, Victoria University, Melbourne, VIC, Australia
| | - Vasiliki Mahairaki
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Richman Family Precision Medicine Center of Excellence in Alzheimer’s Disease, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kenneth W. Witwer
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Richman Family Precision Medicine Center of Excellence in Alzheimer’s Disease, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
25
|
Wang P, Liao H, Wang Q, Xie H, Wang H, Yang M, Liu S. L1 Syndrome Prenatal Diagnosis Supplemented by Functional Analysis of One L1CAM Gene Missense Variant. Reprod Sci 2021; 29:768-780. [PMID: 34914080 PMCID: PMC8863719 DOI: 10.1007/s43032-021-00828-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 12/06/2021] [Indexed: 11/30/2022]
Abstract
L1 syndrome, a complex X-linked neurological disorder, is caused by mutations in the L1 cell adhesion molecule (L1CAM) gene. L1CAM molecule is a member of immunoglobulin (Ig) superfamily of neural cell adhesion molecules (CAMs), which plays a pivotal role in the developing nervous system. In this study, a L1CAM gene exonic missense variant (c.1108G > A, p.G370R) was identified in two induced fetuses (abnormal fetuses), who presented corpus callosum agenesis accompanied with hydrocephalus. Clinical data, published literature, online database, and bioinformatic analysis suggest that the single-nucleotide variant of L1CAM gene is a likely pathogenic mutation. In vitro assays were performed to evaluate the effects of this variant. Based on NSC-34/COS-7 cells transfected with wild-type (L1-WT) and mutated (L1-G370R) plasmids, the L1CAM gene exonic missense variant (c.1108G > A, p.G370R) reduced cell surface expression, induced partial endoplasmic reticulum retention, affected posttranslational modification, and reduced protein’s homophilic adhesive ability, but did not induce endoplasmic reticulum stress, which might probably associate with L1 syndrome. Finally, 35 isolated fetuses were screened for L1CAM gene variants by Sanger sequencing. These cases all prenatally suspected of corpus callosum agenesis accompanied with hydrocephalus, which may relate to L1 syndrome. Consequently, one L1CAM gene single missense variant (c.550C > T, p.R184W) was detected in one fetus. Our results provided evidence that the L1CAM gene missense variant (c.1108G > A, p.G370R) may relate to L1 syndrome. The findings of this study suggest a potential possibility of L1CAM gene screening for prenatal diagnoses for fetuses presented corpus callosum agenesis accompanied with hydrocephalus.
Collapse
Affiliation(s)
- Ping Wang
- Department of Obstetrics & Gynecology, West China Second University Hospital, Sichuan University, No. 20, Section 3, Renminnan Road, Chengdu, 610041, Sichuan, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, Sichuan, China
| | - Hong Liao
- Department of Obstetrics & Gynecology, West China Second University Hospital, Sichuan University, No. 20, Section 3, Renminnan Road, Chengdu, 610041, Sichuan, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, Sichuan, China
| | - Quyou Wang
- Department of Obstetrics & Gynecology, West China Second University Hospital, Sichuan University, No. 20, Section 3, Renminnan Road, Chengdu, 610041, Sichuan, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, Sichuan, China
| | - Hanbing Xie
- Department of Obstetrics & Gynecology, West China Second University Hospital, Sichuan University, No. 20, Section 3, Renminnan Road, Chengdu, 610041, Sichuan, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, Sichuan, China
| | - He Wang
- Department of Obstetrics & Gynecology, West China Second University Hospital, Sichuan University, No. 20, Section 3, Renminnan Road, Chengdu, 610041, Sichuan, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, Sichuan, China
| | - Mei Yang
- Department of Obstetrics & Gynecology, West China Second University Hospital, Sichuan University, No. 20, Section 3, Renminnan Road, Chengdu, 610041, Sichuan, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, Sichuan, China
| | - Shanling Liu
- Department of Obstetrics & Gynecology, West China Second University Hospital, Sichuan University, No. 20, Section 3, Renminnan Road, Chengdu, 610041, Sichuan, China. .,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
26
|
Sung M, Sung SE, Kang KK, Choi JH, Lee S, Kim K, Lim JH, Lee GW, Rim HD, Won S, Kim BS, Kim K, Jang S, Kwak SG, Woo J, Seo MS. Serum-Derived Neuronal Exosomal microRNAs as Stress-Related Biomarkers in an Atopic Dermatitis Model. Biomedicines 2021; 9:biomedicines9121764. [PMID: 34944580 PMCID: PMC8698818 DOI: 10.3390/biomedicines9121764] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 11/13/2021] [Accepted: 11/23/2021] [Indexed: 11/29/2022] Open
Abstract
Chronic allergic inflammatory skin disease—atopic dermatitis (AD)—is characterized by eczema, pruritus, xeroderma, and lichenification. Psychological stress is one cause of this disease; however, psychological stress will also result from the presence of AD symptoms. Previous studies have shown that psychological stress triggers neuroinflammation in the brain, where microRNAs (miRNAs) in the neuronal exosomes (nEVs) were analyzed to identify the composition of the miRNAs in the nEVs and how they were altered by AD. In this study, the AD model was induced by treatment with 2,4-dinitrochlorobenzene (DNCB). The expression patterns of neuroinflammation markers, such as brain-derived neurotrophic factor, cyclooxygenase-2, and glial fibrillary acidic protein, were subsequently evaluated over time. Among these groups, there was a significant difference in DNCB 14 days expression compared with the control; therefore, nEVs were isolated from serum and next-generation sequencing was performed. The results demonstrate that 9 miRNAs were upregulated and 16 were downregulated in the DNCB 14 days compared with the control. Previous studies have shown that some of these miRNAs are associated with stress and stress-induced depression, which suggests that the miRNAs in nEVs may also be stress-related biomarkers.
Collapse
Affiliation(s)
- Minkyoung Sung
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Korea; (M.S.); (S.-E.S.); (K.-K.K.); (J.-H.C.); (S.L.); (K.K.)
- Department of Psychiatry, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (H.-D.R.); (S.W.); (B.-S.K.); (K.K.); (S.J.)
| | - Soo-Eun Sung
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Korea; (M.S.); (S.-E.S.); (K.-K.K.); (J.-H.C.); (S.L.); (K.K.)
| | - Kyung-Ku Kang
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Korea; (M.S.); (S.-E.S.); (K.-K.K.); (J.-H.C.); (S.L.); (K.K.)
| | - Joo-Hee Choi
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Korea; (M.S.); (S.-E.S.); (K.-K.K.); (J.-H.C.); (S.L.); (K.K.)
| | - Sijoon Lee
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Korea; (M.S.); (S.-E.S.); (K.-K.K.); (J.-H.C.); (S.L.); (K.K.)
| | - KilSoo Kim
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Korea; (M.S.); (S.-E.S.); (K.-K.K.); (J.-H.C.); (S.L.); (K.K.)
- College of Veterinary Medicine, Kyungpook National University, 80 Daehakro, Buk-gu, Daegu 41566, Korea
| | - Ju-Hyeon Lim
- New Drug Development Center, Osong Medical Innovation Foundation, Cheongju 28160, Korea;
- Department of Orthopedic Surgery, Yeungnam University Medical Center, Yeungnam University College of Medicine, 170 Hyonchung-ro, Namgu, Daegu 42415, Korea;
| | - Gun Woo Lee
- Department of Orthopedic Surgery, Yeungnam University Medical Center, Yeungnam University College of Medicine, 170 Hyonchung-ro, Namgu, Daegu 42415, Korea;
| | - Hyo-Deog Rim
- Department of Psychiatry, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (H.-D.R.); (S.W.); (B.-S.K.); (K.K.); (S.J.)
| | - Seunghee Won
- Department of Psychiatry, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (H.-D.R.); (S.W.); (B.-S.K.); (K.K.); (S.J.)
| | - Byung-Soo Kim
- Department of Psychiatry, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (H.-D.R.); (S.W.); (B.-S.K.); (K.K.); (S.J.)
| | - Kyungmin Kim
- Department of Psychiatry, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (H.-D.R.); (S.W.); (B.-S.K.); (K.K.); (S.J.)
| | - Seoyoung Jang
- Department of Psychiatry, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (H.-D.R.); (S.W.); (B.-S.K.); (K.K.); (S.J.)
| | - Sang Gyu Kwak
- Department of Medical Statistics, School of Medicine, Catholic University of Daegu, Daegu 42472, Korea;
| | - Jungmin Woo
- Department of Psychiatry, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (H.-D.R.); (S.W.); (B.-S.K.); (K.K.); (S.J.)
- Correspondence: (J.W.); (M.-S.S.); Tel.: +82-200-5747 (J.W.); +82-53-790-5727 (M.-S.S.)
| | - Min-Soo Seo
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Korea; (M.S.); (S.-E.S.); (K.-K.K.); (J.-H.C.); (S.L.); (K.K.)
- Correspondence: (J.W.); (M.-S.S.); Tel.: +82-200-5747 (J.W.); +82-53-790-5727 (M.-S.S.)
| |
Collapse
|
27
|
Scapin G, Gasparotto M, Peterle D, Tescari S, Porcellato E, Piovesan A, Righetto I, Acquasaliente L, De Filippis V, Filippini F. A conserved Neurite Outgrowth and Guidance motif with biomimetic potential in neuronal Cell Adhesion Molecules. Comput Struct Biotechnol J 2021; 19:5622-5636. [PMID: 34712402 PMCID: PMC8529090 DOI: 10.1016/j.csbj.2021.10.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 09/27/2021] [Accepted: 10/03/2021] [Indexed: 01/02/2023] Open
Abstract
The discovery of conserved protein motifs can, in turn, unveil important regulatory signals, and when properly designed, synthetic peptides derived from such motifs can be used as biomimetics for biotechnological and therapeutic purposes. We report here that specific Ig-like repeats from the extracellular domains of neuronal Cell Adhesion Molecules share a highly conserved Neurite Outgrowth and Guidance (NOG) motif, which mediates homo- and heterophilic interactions crucial in neural development and repair. Synthetic peptides derived from the NOG motif of such proteins can boost neuritogenesis, and this potential is also retained by peptides with recombinant sequences, when fitting the NOG sequence pattern. The NOG motif discovery not only provides one more tile to the complex puzzle of neuritogenesis, but also opens the route to new neural regeneration strategies via a tunable biomimetic toolbox.
Collapse
Affiliation(s)
- Giorgia Scapin
- Synthetic Biology and Biotechnology Unit, Department of Biology, University of Padua, 35131, Italy
| | - Matteo Gasparotto
- Synthetic Biology and Biotechnology Unit, Department of Biology, University of Padua, 35131, Italy
| | - Daniele Peterle
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, 35131, Italy
| | - Simone Tescari
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, 35131, Italy
| | - Elena Porcellato
- Synthetic Biology and Biotechnology Unit, Department of Biology, University of Padua, 35131, Italy.,Department of Pharmaceutical and Pharmacological Sciences, University of Padua, 35131, Italy
| | - Alberto Piovesan
- Synthetic Biology and Biotechnology Unit, Department of Biology, University of Padua, 35131, Italy.,Department of Pharmaceutical and Pharmacological Sciences, University of Padua, 35131, Italy
| | - Irene Righetto
- Synthetic Biology and Biotechnology Unit, Department of Biology, University of Padua, 35131, Italy
| | - Laura Acquasaliente
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, 35131, Italy
| | - Vincenzo De Filippis
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, 35131, Italy
| | - Francesco Filippini
- Synthetic Biology and Biotechnology Unit, Department of Biology, University of Padua, 35131, Italy
| |
Collapse
|
28
|
The emerging roles of circular RNAs in vessel co-option and vasculogenic mimicry: clinical insights for anti-angiogenic therapy in cancers. Cancer Metastasis Rev 2021; 41:173-191. [PMID: 34664157 DOI: 10.1007/s10555-021-10000-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 10/12/2021] [Indexed: 12/20/2022]
Abstract
Unexpected resistance to anti-angiogenic treatment prompted the investigation of non-angiogenic tumor processes. Vessel co-option (VC) and vasculogenic mimicry (VM) are recognized as primary non-angiogenic mechanisms. In VC, cancer cells utilize pre-existing blood vessels for support, whereas in VM, cancer cells channel and provide blood flow to rapidly growing tumors. Both processes have been implicated in the development of tumor and resistance to anti-angiogenic drugs in many tumor types. The morphology, but rare molecular alterations have been investigated in VC and VM. There is a pressing need to better understand the underlying cellular and molecular mechanisms. Here, we review the emerging circular RNA (circRNA)-mediated regulation of non-angiogenic processes, VC and VM.
Collapse
|
29
|
Sung M, Sung SE, Kang KK, Choi JH, Lee S, Kim K, Lim JH, Lee GW, Rim HD, Kim BS, Won S, Kim K, Jang S, Seo MS, Woo J. Serum-Derived Neuronal Exosomal miRNAs as Biomarkers of Acute Severe Stress. Int J Mol Sci 2021; 22:9960. [PMID: 34576126 PMCID: PMC8470330 DOI: 10.3390/ijms22189960] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 06/21/2021] [Accepted: 06/23/2021] [Indexed: 12/11/2022] Open
Abstract
Stress is the physical and psychological tension felt by an individual while adapting to difficult situations. Stress is known to alter the expression of stress hormones and cause neuroinflammation in the brain. In this study, miRNAs in serum-derived neuronal exosomes (nEVs) were analyzed to determine whether differentially expressed miRNAs could be used as biomarkers of acute stress. Specifically, acute severe stress was induced in Sprague-Dawley rats via electric foot-shock treatment. In this acute severe-stress model, time-dependent changes in the expression levels of stress hormones and neuroinflammation-related markers were analyzed. In addition, nEVs were isolated from the serum of control mice and stressed mice at various time points to determine when brain damage was most prominent; this was found to be 7 days after foot shock. Next-generation sequencing was performed to compare neuronal exosomal miRNA at day 7 with the neuronal exosomal miRNA of the control group. From this analysis, 13 upregulated and 11 downregulated miRNAs were detected. These results show that specific miRNAs are differentially expressed in nEVs from an acute severe-stress animal model. Thus, this study provides novel insights into potential stress-related biomarkers.
Collapse
Affiliation(s)
- Minkyoung Sung
- Department of Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Korea; (M.S.); (S.-E.S.); (K.-K.K.); (J.-H.C.); (S.L.); (K.K.)
- Department of Psychiatry, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (H.-D.R.); (B.-S.K.); (S.W.); (K.K.); (S.J.)
| | - Soo-Eun Sung
- Department of Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Korea; (M.S.); (S.-E.S.); (K.-K.K.); (J.-H.C.); (S.L.); (K.K.)
| | - Kyung-Ku Kang
- Department of Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Korea; (M.S.); (S.-E.S.); (K.-K.K.); (J.-H.C.); (S.L.); (K.K.)
| | - Joo-Hee Choi
- Department of Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Korea; (M.S.); (S.-E.S.); (K.-K.K.); (J.-H.C.); (S.L.); (K.K.)
| | - Sijoon Lee
- Department of Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Korea; (M.S.); (S.-E.S.); (K.-K.K.); (J.-H.C.); (S.L.); (K.K.)
| | - KilSoo Kim
- Department of Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Korea; (M.S.); (S.-E.S.); (K.-K.K.); (J.-H.C.); (S.L.); (K.K.)
- Department of Veterinary Toxicology, College of Veterinary Medicine, Kyungpook National University, 80 Daehakro, Buk-gu, Daegu 41566, Korea
| | - Ju-Hyeon Lim
- New Drug Development Center, Osong Medical Innovation Foundation, Chungbuk 28160, Korea; (J.-H.L.); (G.W.L.)
- Department of Orthopedic Surgery, Yeungnam University College of Medicine, Yeungnam University Medical Center, 170 Hyonchung-ro, Namgu, Daegu 42415, Korea
| | - Gun Woo Lee
- Department of Orthopedic Surgery, Yeungnam University College of Medicine, Yeungnam University Medical Center, 170 Hyonchung-ro, Namgu, Daegu 42415, Korea
| | - Hyo-Deog Rim
- Department of Psychiatry, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (H.-D.R.); (B.-S.K.); (S.W.); (K.K.); (S.J.)
| | - Byung-Soo Kim
- Department of Psychiatry, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (H.-D.R.); (B.-S.K.); (S.W.); (K.K.); (S.J.)
| | - Seunghee Won
- Department of Psychiatry, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (H.-D.R.); (B.-S.K.); (S.W.); (K.K.); (S.J.)
| | - Kyungmin Kim
- Department of Psychiatry, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (H.-D.R.); (B.-S.K.); (S.W.); (K.K.); (S.J.)
| | - Seoyoung Jang
- Department of Psychiatry, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (H.-D.R.); (B.-S.K.); (S.W.); (K.K.); (S.J.)
| | - Min-Soo Seo
- Department of Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Korea; (M.S.); (S.-E.S.); (K.-K.K.); (J.-H.C.); (S.L.); (K.K.)
| | - Jungmin Woo
- Department of Psychiatry, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (H.-D.R.); (B.-S.K.); (S.W.); (K.K.); (S.J.)
| |
Collapse
|
30
|
Dräger O, Metz K, Busch M, Dünker N. Role of L1CAM in retinoblastoma tumorigenesis: identification of novel therapeutic targets. Mol Oncol 2021; 16:957-981. [PMID: 34228897 PMCID: PMC8847994 DOI: 10.1002/1878-0261.13054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 05/11/2021] [Accepted: 07/05/2021] [Indexed: 11/08/2022] Open
Abstract
The study presented focuses on the role of the neuronal cell adhesion molecule L1 cell adhesion molecule (L1CAM) in retinoblastoma (RB), the most common malignant intraocular childhood tumor. L1CAM is differentially expressed in a variety of human cancers and has been suggested as a promising therapeutic target. We likewise observed differential expression patterns for L1CAM in RB cell lines and patient samples. The two proteases involved in ectodomain shedding of L1CAM (L1CAM sheddases: ADAM10 and ADAM17) were likewise differentially expressed in the RB cell lines investigated, and an involvement in L1CAM processing in RB cells could be verified. We also identified ezrin, galectin-3, and fibroblast growth factor basic as L1CAM signaling target genes in RB cells. Lentiviral L1CAM knockdown induced apoptosis and reduced cell viability, proliferation, growth, and colony formation capacity of RB cells, whereas L1CAM-overexpressing RB cells displayed the opposite effects. Chicken chorioallantoic membrane assays revealed that L1CAM depletion decreases the tumorigenic and migration potential of RB cells in vivo. Moreover, L1CAM depletion decreased viability and tumor growth of etoposide-resistant RB cell lines upon etoposide treatment in vitro and in vivo. Thus, L1CAM and its processing sheddases are potential novel targets for future therapeutic RB approaches.
Collapse
Affiliation(s)
- Oliver Dräger
- Institute of Anatomy II, Department of Neuroanatomy, University of Duisburg-Essen, Medical Faculty, Germany
| | - Klaus Metz
- Institute of Pathology, University of Duisburg-Essen, Medical Faculty, Germany
| | - Maike Busch
- Institute of Anatomy II, Department of Neuroanatomy, University of Duisburg-Essen, Medical Faculty, Germany
| | - Nicole Dünker
- Institute of Anatomy II, Department of Neuroanatomy, University of Duisburg-Essen, Medical Faculty, Germany
| |
Collapse
|
31
|
Bone Marrow-Derived SH-SY5Y Neuroblastoma Cells Infected with Kaposi's Sarcoma-Associated Herpesvirus Display Unique Infection Phenotypes and Growth Properties. J Virol 2021; 95:e0000321. [PMID: 33853962 DOI: 10.1128/jvi.00003-21] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) is an important oncogenic virus previously shown to be neurotropic, but studies on neuronal cell infection and pathogenesis are still very limited. Here, we characterized the effects of KSHV infection on neuronal SH-SY5Y cells by the recombinant virus rKSHV.219, which expresses both green fluorescent protein (GFP) and red fluorescent protein (RFP) to reflect the latent and lytic phases of infection. We demonstrated that infected cells have a higher growth rate and that KSHV infection can be sustained. Interestingly, the infected cells can transition spontaneously back and forth between lytic and latent phases of infection, producing progeny viruses but without any adverse effects on cell growth. In addition, transcriptome analysis of viral and cellular genes in latent and lytic cells showed that unlike other infected cell lines, the latently infected cells expressed both latent and most, but not all, of the lytic genes required for infectious virion production. The viral genes uniquely expressed by the lytic cells were mainly involved in the early steps of virus binding. Some of the cellular genes that were deregulated in both latently and lytically infected cells are involved in cell adhesion, cell signal pathways, and tumorigenesis. The downregulated cellular CCDN1, PAX5, and NFASC and upregulated CTGF, BMP4, YAP1, LEF1, and HLA-DRB1 genes were found to be associated with cell adhesion molecules (CAMs), hippo signaling, and cancer. These deregulated genes may be involved in creating an environment that is unique in neuronal cells to sustain cell growth upon KSHV infection and not observed in other infected cell types. IMPORTANCE Our study has provided evidence that neuronal SH-SY5Y cells displayed unique cellular responses upon KSHV infection. Unlike other infected cells, this neuronal cell line displayed a higher growth rate upon infection and can spontaneously transition back and forth between latent and lytic phases of infection. Unlike other latently infected cells, a number of lytic genes were also expressed in the latent phase of infection in addition to the established latent viral genes. They may play a role in deregulating a number of host genes that are involved in cell signaling and tumorigenesis in order to sustain the infection and growth advantages for the cells. Our study has provided novel insights into KSHV infection of neuronal cells and a potential new model for further studies to explore the underlying mechanism in viral and host interactions for neuronal cells and the association of KSHV with neuronal diseases.
Collapse
|
32
|
Nikulin S, Zakharova G, Poloznikov A, Raigorodskaya M, Wicklein D, Schumacher U, Nersisyan S, Bergquist J, Bakalkin G, Astakhova L, Tonevitsky A. Effect of the Expression of ELOVL5 and IGFBP6 Genes on the Metastatic Potential of Breast Cancer Cells. Front Genet 2021; 12:662843. [PMID: 34149804 PMCID: PMC8206645 DOI: 10.3389/fgene.2021.662843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 04/20/2021] [Indexed: 12/09/2022] Open
Abstract
Breast cancer (BC) is the leading cause of death from malignant neoplasms among women worldwide, and metastatic BC presents the biggest problems for treatment. Previously, it was shown that lower expression of ELOVL5 and IGFBP6 genes is associated with a higher risk of the formation of distant metastases in BC. In this work, we studied the change in phenotypical traits, as well as in the transcriptomic and proteomic profiles of BC cells as a result of the stable knockdown of ELOVL5 and IGFBP6 genes. The knockdown of ELOVL5 and IGFBP6 genes was found to lead to a strong increase in the expression of the matrix metalloproteinase (MMP) MMP1. These results were in good agreement with the correlation analysis of gene expression in tumor samples from patients and were additionally confirmed by zymography. The knockdown of ELOVL5 and IGFBP6 genes was also discovered to change the expression of a group of genes involved in the formation of intercellular contacts. In particular, the expression of the CDH11 gene was markedly reduced, which also complies with the correlation analysis. The spheroid formation assay showed that intercellular adhesion decreased as a result of the knockdown of the ELOVL5 and IGFBP6 genes. Thus, the obtained data indicate that malignant breast tumors with reduced expression of the ELOVL5 and IGFBP6 genes can metastasize with a higher probability due to a more efficient invasion of tumor cells.
Collapse
Affiliation(s)
- Sergey Nikulin
- Faculty of Biology and Biotechnologies, National Research University Higher School of Economics, Moscow, Russia
| | | | - Andrey Poloznikov
- Faculty of Biology and Biotechnologies, National Research University Higher School of Economics, Moscow, Russia
- School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia
| | - Maria Raigorodskaya
- Faculty of Biology and Biotechnologies, National Research University Higher School of Economics, Moscow, Russia
- Scientific Research Centre Bioclinicum, Moscow, Russia
| | - Daniel Wicklein
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Udo Schumacher
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stepan Nersisyan
- Faculty of Biology and Biotechnologies, National Research University Higher School of Economics, Moscow, Russia
| | - Jonas Bergquist
- Department of Chemistry – BMC, Uppsala University, Uppsala, Sweden
| | - Georgy Bakalkin
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Lidiia Astakhova
- Scientific Research Centre Bioclinicum, Moscow, Russia
- School of Life Sciences, Immanuel Kant Baltic Federal University, Kaliningrad, Russia
| | - Alexander Tonevitsky
- Faculty of Biology and Biotechnologies, National Research University Higher School of Economics, Moscow, Russia
- Laboratory of Microfluidic Technologies for Biomedicine, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, Moscow, Russia
| |
Collapse
|
33
|
Del Favero G, Zeugswetter M, Kiss E, Marko D. Endoplasmic Reticulum Adaptation and Autophagic Competence Shape Response to Fluid Shear Stress in T24 Bladder Cancer Cells. Front Pharmacol 2021; 12:647350. [PMID: 34012396 PMCID: PMC8126838 DOI: 10.3389/fphar.2021.647350] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 02/17/2021] [Indexed: 12/26/2022] Open
Abstract
Accumulation of xenobiotics and waste metabolites in the urinary bladder is constantly accompanied by shear stress originating from the movement of the luminal fluids. Hence, both chemical and physical cues constantly modulate the cellular response in health and disease. In line, bladder cells have to maintain elevated mechanosensory competence together with chemical stress response adaptation potential. However, much of the molecular mechanisms sustaining this plasticity is currently unknown. Taking this as a starting point, we investigated the response of T24 urinary bladder cancer cells to shear stress comparing morphology to functional performance. T24 cells responded to the shear stress protocol (flow speed of 0.03 ml/min, 3 h) by significantly increasing their surface area. When exposed to deoxynivalenol-3-sulfate (DON-3-Sulf), bladder cells increased this response in a concentration-dependent manner (0.1-1 µM). DON-3-Sulf is a urinary metabolite of a very common food contaminant mycotoxin (deoxynivalenol, DON) and was already described to enhance proliferation of cancer cells. Incubation with DON-3-Sulf also caused the enlargement of the endoplasmic reticulum (ER), decreased the lysosomal movement, and increased the formation of actin stress fibers. Similar remodeling of the endoplasmic reticulum and area spread after shear stress were observed upon incubation with the autophagy activator rapamycin (1-100 nM). Performance of experiments in the presence of chloroquine (chloroquine, 30 μM) further contributed to shed light on the mechanistic link between adaptation to the biomechanical stimulation and ER stress response. At the molecular level, we observed that ER reshaping was linked to actin organization, with the two components mutually regulating each other. Indeed, we identified in the ER stress-cytoskeletal rearrangement an important axis defining the physical/chemical response potential of bladder cells and created a workflow for further investigation of urinary metabolites, food constituents, and contaminants, as well as for pharmacological profiling.
Collapse
Affiliation(s)
- Giorgia Del Favero
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Vienna, Austria.,Core Facility Multimodal Imaging, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Michael Zeugswetter
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Endre Kiss
- Core Facility Multimodal Imaging, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Doris Marko
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Vienna, Austria
| |
Collapse
|
34
|
Srinivasamurthy M, Kakanahalli N, Benakanal SV. A truncation mutation in the <i>L1CAM</i> gene in a child with hydrocephalus. AIMS MOLECULAR SCIENCE 2021. [DOI: 10.3934/molsci.2021017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
<abstract>
<p>Hydrocephalus is a neurodevelopmental, X-linked recessive disorder caused by mutations in the <italic>L1CAM</italic> gene. The <italic>L1CAM</italic> gene encodes for L1CAM protein which is essential for the nervous system development including adhesion between neurons, Myelination, Synaptogenesis etc. Herein, the present study has reported mutations in L1 syndrome patient with Hydrocephalus and Adducted thumb. Genomic DNA was extracted from patients whole blood (n = 18). The 11 exons of the <italic>L1CAM</italic> gene were amplified using specific PCR primers. The sequenced data was analysed and the pathogenicity of the mutation was predicted using the various bioinformatics programs: PROVEAN, PolyPhen2, and MUpro. The results revealed that the proband described here had nonsense mutation G1120→T at position 1120 in exon 9 which is in extracellular immunoglobulin domain (Ig4) of the <italic>L1CAM</italic> gene. This nonsense mutation is found to be truncated with a deleterious effect on developing brain of the child, and this is the first report of this novel mutation in patient with X-linked Hydrocephalus in India.</p>
</abstract>
Collapse
|
35
|
McDonald D, Wu Y, Dailamy A, Tat J, Parekh U, Zhao D, Hu M, Tipps A, Zhang K, Mali P. Defining the Teratoma as a Model for Multi-lineage Human Development. Cell 2020; 183:1402-1419.e18. [PMID: 33152263 PMCID: PMC7704916 DOI: 10.1016/j.cell.2020.10.018] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Revised: 06/06/2020] [Accepted: 10/09/2020] [Indexed: 12/14/2022]
Abstract
We propose that the teratoma, a recognized standard for validating pluripotency in stem cells, could be a promising platform for studying human developmental processes. Performing single-cell RNA sequencing (RNA-seq) of 179,632 cells across 23 teratomas from 4 cell lines, we found that teratomas reproducibly contain approximately 20 cell types across all 3 germ layers, that inter-teratoma cell type heterogeneity is comparable with organoid systems, and teratoma gut and brain cell types correspond well to similar fetal cell types. Furthermore, cellular barcoding confirmed that injected stem cells robustly engraft and contribute to all lineages. Using pooled CRISPR-Cas9 knockout screens, we showed that teratomas can enable simultaneous assaying of the effects of genetic perturbations across all germ layers. Additionally, we demonstrated that teratomas can be sculpted molecularly via microRNA (miRNA)-regulated suicide gene expression to enrich for specific tissues. Taken together, teratomas are a promising platform for modeling multi-lineage development, pan-tissue functional genetic screening, and tissue engineering.
Collapse
Affiliation(s)
- Daniella McDonald
- Department of Bioengineering, University of California, San Diego, San Diego, CA 92093, USA; Biomedical Sciences Graduate Program, University of California, San Diego, San Diego, CA 92093, USA
| | - Yan Wu
- Department of Bioengineering, University of California, San Diego, San Diego, CA 92093, USA
| | - Amir Dailamy
- Department of Bioengineering, University of California, San Diego, San Diego, CA 92093, USA
| | - Justin Tat
- Department of Biological Sciences, University of California, San Diego, San Diego, CA 92093, USA
| | - Udit Parekh
- Department of Electrical and Computer Engineering, University of California, San Diego, San Diego, CA 92093, USA
| | - Dongxin Zhao
- Department of Bioengineering, University of California, San Diego, San Diego, CA 92093, USA
| | - Michael Hu
- Department of Bioengineering, University of California, San Diego, San Diego, CA 92093, USA
| | - Ann Tipps
- School of Medicine, University of California, San Diego, San Diego, CA 92103, USA
| | - Kun Zhang
- Department of Bioengineering, University of California, San Diego, San Diego, CA 92093, USA; Biomedical Sciences Graduate Program, University of California, San Diego, San Diego, CA 92093, USA.
| | - Prashant Mali
- Department of Bioengineering, University of California, San Diego, San Diego, CA 92093, USA; Biomedical Sciences Graduate Program, University of California, San Diego, San Diego, CA 92093, USA.
| |
Collapse
|
36
|
McCann TS, Parrish JK, Hsieh J, Sechler M, Sobral LM, Self C, Jones KL, Goodspeed A, Costello JC, Jedlicka P. KDM5A and PHF2 positively control expression of pro-metastatic genes repressed by EWS/Fli1, and promote growth and metastatic properties in Ewing sarcoma. Oncotarget 2020; 11:3818-3831. [PMID: 33196691 PMCID: PMC7597412 DOI: 10.18632/oncotarget.27737] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 08/24/2020] [Indexed: 02/07/2023] Open
Abstract
Ewing sarcoma is an aggressive malignant neoplasm with high propensity for metastasis and poor clinical outcomes. The EWS/Fli1 oncofusion is the disease driver in > 90% of cases, but presents a difficult therapeutic target. Moreover, EWS/Fli1 plays a complex role in disease progression, with inhibitory effects on critical steps of metastasis. Like many other pediatric cancers, Ewing sarcoma is a disease marked by epigenetic dysregulation. Epigenetic mechanisms present alternative targeting opportunities, but their contributions to Ewing sarcoma metastasis and disease progression remain poorly understood. Here, we show that the epigenetic regulators KDM5A and PHF2 promote growth and metastatic properties in Ewing sarcoma, and, strikingly, activate expression many pro-metastatic genes repressed by EWS/Fli1. These genes include L1CAM, which is associated with adverse outcomes in Ewing sarcoma, and promotes migratory and invasive properties. KDM5A and PHF2 retain their growth promoting effects in more metastatically potent EWS/Fli1low cells, and PHF2 promotes both invasion and L1CAM expression in this cell population. Furthermore, KDM5A and PHF2 each contribute to the increased metastatic potency of EWS/Fli1low cells in vivo. Together, these studies identify KDM5A and PHF2 as novel disease-promoting factors, and potential new targets, in Ewing sarcoma, including the more metastatically potent EWS/Fli1low cell population.
Collapse
Affiliation(s)
- Tyler S McCann
- Department of Pathology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Janet K Parrish
- Department of Pathology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Joseph Hsieh
- Department of Pathology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA.,Medical Scientist Training Program, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA.,Cancer Biology Graduate Program, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Marybeth Sechler
- Department of Pathology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA.,Cancer Biology Graduate Program, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Lays M Sobral
- Department of Pathology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Chelsea Self
- Department of Pediatrics, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Kenneth L Jones
- Department of Pediatrics, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA.,Bioinformatics Shared Resource, University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, CO, USA
| | - Andrew Goodspeed
- Department of Pharmacology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA.,Bioinformatics Shared Resource, University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, CO, USA
| | - James C Costello
- Department of Pharmacology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA.,Bioinformatics Shared Resource, University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, CO, USA
| | - Paul Jedlicka
- Department of Pathology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA.,Medical Scientist Training Program, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA.,Cancer Biology Graduate Program, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
37
|
Transcriptional analysis of cleft palate in TGFβ3 mutant mice. Sci Rep 2020; 10:14940. [PMID: 32913205 PMCID: PMC7483747 DOI: 10.1038/s41598-020-71636-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 08/17/2020] [Indexed: 12/30/2022] Open
Abstract
Cleft palate (CP) is one of the most common craniofacial birth defects, impacting about 1 in 800 births in the USA. Tgf-β3 plays a critical role in regulating murine palate development, and Tgf-β3 null mutants develop cleft palate with 100% penetrance. In this study, we compared global palatal transcriptomes of wild type (WT) and Tgf-β3 −/− homozygous (HM) mouse embryos at the crucial palatogenesis stages of E14.5, and E16.5, using RNA-seq data. We found 1,809 and 2,127 differentially expressed genes at E16.5 vs. E14.5 in the WT and HM groups, respectively (adjusted p < 0.05; |fold change|> 2.0). We focused on the genes that were uniquely up/downregulated in WT or HM at E16.5 vs. E14.5 to identify genes associated with CP. Systems biology analysis relating to cell behaviors and function of WT and HM specific genes identified functional non-Smad pathways and preference of apoptosis to epithelial-mesenchymal transition. We identified 24 HM specific and 11 WT specific genes that are CP-related and/or involved in Tgf-β3 signaling. We validated the expression of 29 of the 35 genes using qRT-PCR and the trend of mRNA expression is similar to that of RNA-seq data . Our results enrich our understanding of genes associated with CP that are directly or indirectly regulated via TGF-β.
Collapse
|
38
|
Israfil A, Israfil N. RETRACTED: Temperament gene inheritance. Meta Gene 2020. [DOI: 10.1016/j.mgene.2020.100728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
39
|
Ghaemmaghami AB, Mahjoubin-Tehran M, Movahedpour A, Morshedi K, Sheida A, Taghavi SP, Mirzaei H, Hamblin MR. Role of exosomes in malignant glioma: microRNAs and proteins in pathogenesis and diagnosis. Cell Commun Signal 2020; 18:120. [PMID: 32746854 PMCID: PMC7397575 DOI: 10.1186/s12964-020-00623-9] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 07/02/2020] [Indexed: 02/07/2023] Open
Abstract
Malignant gliomas are the most common and deadly type of central nervous system tumors. Despite some advances in treatment, the mean survival time remains only about 1.25 years. Even after surgery, radiotherapy and chemotherapy, gliomas still have a poor prognosis. Exosomes are the most common type of extracellular vesicles with a size range of 30 to 100 nm, and can act as carriers of proteins, RNAs, and other bioactive molecules. Exosomes play a key role in tumorigenesis and resistance to chemotherapy or radiation. Recent evidence has shown that exosomal microRNAs (miRNAs) can be detected in the extracellular microenvironment, and can also be transferred from cell to cell via exosome secretion and uptake. Therefore, many recent studies have focused on exosomal miRNAs as important cellular regulators in various physiological and pathological conditions. A variety of exosomal miRNAs have been implicated in the initiation and progression of gliomas, by activating and/or inhibiting different signaling pathways. Exosomal miRNAs could be used as therapeutic agents to modulate different biological processes in gliomas. Exosomal miRNAs derived from mesenchymal stem cells could also be used for glioma treatment. The present review summarizes the exosomal miRNAs that have been implicated in the pathogenesis, diagnosis and treatment of gliomas. Moreover, exosomal proteins could also be involved in glioma pathogenesis. Exosomal miRNAs and proteins could also serve as non-invasive biomarkers for prognosis and disease monitoring. Video Abstract.
Collapse
Affiliation(s)
- Amir B. Ghaemmaghami
- grid.17063.330000 0001 2157 2938Department of Psychology, Behaviour, Genetics and Neurobiology Program, University of Toronto, Toronto, Canada
| | - Maryam Mahjoubin-Tehran
- grid.411583.a0000 0001 2198 6209Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran ,grid.411583.a0000 0001 2198 6209Department of Medical Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ahmad Movahedpour
- grid.412571.40000 0000 8819 4698Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran ,grid.412571.40000 0000 8819 4698Student research committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Korosh Morshedi
- grid.444768.d0000 0004 0612 1049School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Amirhossein Sheida
- grid.444768.d0000 0004 0612 1049School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Seyed Pouya Taghavi
- grid.444768.d0000 0004 0612 1049School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Hamed Mirzaei
- grid.444768.d0000 0004 0612 1049Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Michael R. Hamblin
- grid.38142.3c000000041936754XWellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, USA ,grid.412988.e0000 0001 0109 131XLaser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, Johannesburg, 2028 South Africa
| |
Collapse
|
40
|
Aharon A, Spector P, Ahmad RS, Horrany N, Sabbach A, Brenner B, Aharon-Peretz J. Extracellular Vesicles of Alzheimer's Disease Patients as a Biomarker for Disease Progression. Mol Neurobiol 2020; 57:4156-4169. [PMID: 32676990 DOI: 10.1007/s12035-020-02013-1] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Accepted: 07/08/2020] [Indexed: 12/23/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative brain pathology and the most common form of dementia. Evidence suggests that extracellular vesicles (EVs) containing cytokines and microRNA are involved in inflammation regulation. The current study aimed to explore a potential impact of AD patients' EVs on disease progression. Blood samples were collected after obtaining signed informed consent (No. 0462-14-RMB) from 42 AD patients at three stages of disease severity and from 19 healthy controls (HC). EV size and concentration were studied by nanotracking analysis. EV membrane antigens were defined by flow cytometry and Western blot; EV protein contents were screened by protein array; the miRNA content was screened by nanostring technology and validated by RT-PCR. HC and AD patients' EVs consisted of a mixture of small (< 100 nm) and larger vesicles. The myelin oligodendrocyte glycoprotein (MOG) expression on EVs correlated with disease severity. EVs of patients with moderate and severe AD had significantly higher levels of MOG, compared with mild AD patients. Levels of EVs expressing the axonal glycoprotein CD171 were significantly higher in severe AD patients than in HC. Increase in endothelial EVs was observed in AD patients. An above twofold increase was found in the content of inflammatory cytokines and > 50% decrease in growth factors in AD patients' EVs compared with HC-EVs. Levels of let-7g-5p, miR126-3p, miR142-3p, miR-146a-5p, and mir223-3p correlated with disease severity. Neural damage, specific miRNA downregulation, and inflammatory cytokine upregulation, found in patients' EVs, might be used as a biomarker reflecting AD severity.
Collapse
Affiliation(s)
- Anat Aharon
- Tel Aviv Sourasky Medical Center, Tel-Aviv, Israel. .,Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel. .,Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel.
| | - Polina Spector
- Cognitive Neurology Unit, Rambam Health Care Campus, Haifa, Israel
| | | | - Nizar Horrany
- Cognitive Neurology Unit, Rambam Health Care Campus, Haifa, Israel
| | - Annie Sabbach
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel
| | - Benjamin Brenner
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel.,Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Judith Aharon-Peretz
- Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel.,Cognitive Neurology Unit, Rambam Health Care Campus, Haifa, Israel
| |
Collapse
|
41
|
Chu LY, Peng YH, Yang T, Fang WK, Hong CQ, Huang LS, Xu LY, Li EM, Xu YW, Xie JJ. Circulating Levels of L1-cell Adhesion Molecule as a Serum Biomarker for Early Detection of Gastric Cancer and Esophagogastric Junction Adenocarcinoma. J Cancer 2020; 11:5395-5402. [PMID: 32742486 PMCID: PMC7391208 DOI: 10.7150/jca.41100] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 04/21/2020] [Indexed: 02/05/2023] Open
Abstract
Background: Low serum L1 cell adhesion molecule (L1CAM) has been found in several malignant tumors. Here, we aimed to evaluate the diagnostic potential for serum L1CAM in patients with gastric cancers (GC) and esophagogastric junction adenocarcinoma (EJA). Methods: Enzyme-linked immunosorbent assay (ELISA) was carried out to detect L1CAM level in sera of 148 GC patients, 59 EJA patients and 148 healthy controls. Receiver operating characteristics (ROC) was employed to evaluate diagnostic accuracy. Results: The concentrations of serum L1CAM were significantly lower in GC and EJA than those in healthy controls (P<0.001). Detection of L1CAM provided a sensitivity of 83.1%, a specificity of 62.2%, and an area under the curve (AUC) of 0.769 (95% CI: 0.715-0.823) in diagnosing GC, and a sensitivity of 66.1%, a specificity of 62.2%, and an AUC of 0.672 (95% CI: 0.590-0.755) in diagnosing EJA. Similar results were observed in the diagnosis of early-stage GC (0.681 (95%CI: 0.596-0.766)) and early-stage EJA (0.674 (95%CI: 0.528-0.820)). Analysis of clinical data showed that the levels of L1CAM were significantly associated with lymph node metastasis in GC (P<0.05). Conclusions: Our study showed that serum L1CAM might be a diagnostic biomarker for GC and EJA.
Collapse
Affiliation(s)
- Ling-Yu Chu
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, China
| | - Yu-Hui Peng
- Department of Clinical Laboratory Medicine, the Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Tian Yang
- Department of Gastrointestinal Surgery, the First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Wang-Kai Fang
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, China
| | - Chao-Qun Hong
- Department of Oncological Laboratory Research, the Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Li-Sheng Huang
- Department of Radiation Oncology, the Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Li-Yan Xu
- Institute of Oncologic Pathology, Shantou University Medical College, Shantou, China
| | - En-Min Li
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, China
| | - Yi-Wei Xu
- Department of Clinical Laboratory Medicine, the Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Jian-Jun Xie
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, China
| |
Collapse
|
42
|
Maltseva D, Raygorodskaya M, Knyazev E, Zgoda V, Tikhonova O, Zaidi S, Nikulin S, Baranova A, Turchinovich A, Rodin S, Tonevitsky A. Knockdown of the α5 laminin chain affects differentiation of colorectal cancer cells and their sensitivity to chemotherapy. Biochimie 2020; 174:107-116. [PMID: 32334043 DOI: 10.1016/j.biochi.2020.04.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Revised: 04/13/2020] [Accepted: 04/14/2020] [Indexed: 02/07/2023]
Abstract
The interaction of tumor cells with the extracellular matrix (ECM) may affect the rate of cancer progression and metastasis. One of the major components of ECM are laminins, the heterotrimeric glycoproteins consisting of α-, β-, and γ-chains (αβγ). Laminins interact with their cell surface receptors and, thus, regulate multiple cellular processes. In this work, we demonstrate that shRNA-mediated knockdown of the α5 laminin chain results in Wnt- and mTORC1-dependent partial dedifferentiation of colorectal cancer cells. Furthermore, we showed that this dedifferentiation involved activation of ER-stress signaling, pathway promoting the sensitivity of cells to 5-fluorouracil.
Collapse
Affiliation(s)
- Diana Maltseva
- Faculty of Biology and Biotechnology, National Research University Higher School of Economics, Myasnitskaya str. 13/4, 117997, Moscow, Russia; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Miklukho-Maklaya str. 16/10, 117997, Moscow, Russia; Scientific Research Center Bioclinicum, Ugreshskaya str. 2/85, 115088, Moscow, Russia.
| | - Maria Raygorodskaya
- Scientific Research Center Bioclinicum, Ugreshskaya str. 2/85, 115088, Moscow, Russia
| | - Evgeny Knyazev
- Faculty of Biology and Biotechnology, National Research University Higher School of Economics, Myasnitskaya str. 13/4, 117997, Moscow, Russia; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Miklukho-Maklaya str. 16/10, 117997, Moscow, Russia
| | - Victor Zgoda
- Institute of Biomedical Chemistry, Pogodinskaya str. 10, 119121, Moscow, Russia
| | - Olga Tikhonova
- Institute of Biomedical Chemistry, Pogodinskaya str. 10, 119121, Moscow, Russia
| | - Shan Zaidi
- School of Systems Biology, George Mason University, Fairfax, VA, 22030, USA
| | - Sergey Nikulin
- Faculty of Biology and Biotechnology, National Research University Higher School of Economics, Myasnitskaya str. 13/4, 117997, Moscow, Russia; Moscow Institute of Physics and Technology, Institutskiy per. 9, 141700, Dolgoprudny, Russia
| | - Ancha Baranova
- School of Systems Biology, George Mason University, Fairfax, VA, 22030, USA; Moscow Institute of Physics and Technology, Institutskiy per. 9, 141700, Dolgoprudny, Russia; Research Center of Medical Genetics, Moskvorechye str. 1, 115522, Moscow, Russia
| | | | - Sergey Rodin
- Department of Surgical Sciences, Ångström Laboratory, Uppsala University, 752 37, Uppsala, Sweden
| | - Alexander Tonevitsky
- Faculty of Biology and Biotechnology, National Research University Higher School of Economics, Myasnitskaya str. 13/4, 117997, Moscow, Russia; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Miklukho-Maklaya str. 16/10, 117997, Moscow, Russia; Scientific Research Center Bioclinicum, Ugreshskaya str. 2/85, 115088, Moscow, Russia.
| |
Collapse
|
43
|
Rigby MJ, Gomez TM, Puglielli L. Glial Cell-Axonal Growth Cone Interactions in Neurodevelopment and Regeneration. Front Neurosci 2020; 14:203. [PMID: 32210757 PMCID: PMC7076157 DOI: 10.3389/fnins.2020.00203] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 02/24/2020] [Indexed: 12/19/2022] Open
Abstract
The developing nervous system is a complex yet organized system of neurons, glial support cells, and extracellular matrix that arranges into an elegant, highly structured network. The extracellular and intracellular events that guide axons to their target locations have been well characterized in many regions of the developing nervous system. However, despite extensive work, we have a poor understanding of how axonal growth cones interact with surrounding glial cells to regulate network assembly. Glia-to-growth cone communication is either direct through cellular contacts or indirect through modulation of the local microenvironment via the secretion of factors or signaling molecules. Microglia, oligodendrocytes, astrocytes, Schwann cells, neural progenitor cells, and olfactory ensheathing cells have all been demonstrated to directly impact axon growth and guidance. Expanding our understanding of how different glial cell types directly interact with growing axons throughout neurodevelopment will inform basic and clinical neuroscientists. For example, identifying the key cellular players beyond the axonal growth cone itself may provide translational clues to develop therapeutic interventions to modulate neuron growth during development or regeneration following injury. This review will provide an overview of the current knowledge about glial involvement in development of the nervous system, specifically focusing on how glia directly interact with growing and maturing axons to influence neuronal connectivity. This focus will be applied to the clinically-relevant field of regeneration following spinal cord injury, highlighting how a better understanding of the roles of glia in neurodevelopment can inform strategies to improve axon regeneration after injury.
Collapse
Affiliation(s)
- Michael J Rigby
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, United States.,Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, United States.,Waisman Center, University of Wisconsin-Madison, Madison, WI, United States
| | - Timothy M Gomez
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, United States.,Department of Neuroscience, University of Wisconsin-Madison, Madison, WI, United States
| | - Luigi Puglielli
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, United States.,Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, United States.,Waisman Center, University of Wisconsin-Madison, Madison, WI, United States.,Geriatric Research Education Clinical Center, Veterans Affairs Medical Center, Madison, WI, United States
| |
Collapse
|
44
|
He X, Lei S, Zhang Q, Ma L, Li N, Wang J. Deregulation of cell adhesion molecules is associated with progression and poor outcomes in endometrial cancer: Analysis of The Cancer Genome Atlas data. Oncol Lett 2020; 19:1906-1914. [PMID: 32194686 PMCID: PMC7039152 DOI: 10.3892/ol.2020.11295] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 11/15/2019] [Indexed: 01/14/2023] Open
Abstract
Cell adhesion molecules (CAMs) determine the behavior of cancer cells during metastasis. Although some CAMs are dysregulated in certain types of cancer and are associated with cancer progression, to the best of our knowledge, a comprehensive study of CAMs has not been undertaken, particularly in endometrial cancer (EC). In the present study the expression of 225 CAMs in EC patients with various clinicopathological phenotypes were evaluated by statistical analysis using publicly available data from The Cancer Genome Atlas database. The Kaplan-Meier method, and univariate and multivariate Cox proportional hazards regression models were used for survival analyses. Among the differentially expressed CAMs that were associated with aggressive clinicopathological phenotypes, 10 CAM genes were independent prognostic factors compared with other clinicopathological prognostic factors, including stage, grade, age, lymph node status, peritoneal cytology and histological subtype. A total of six genes (L1 cell adhesion molecule, mucin 15, cell surface associated, cell adhesion associated, oncogene regulated, immunoglobulin superfamily member 9B, protocadherin 9 and protocadherin β1) were selected for integrative analysis. The six-gene signature was demonstrated to be an independent prognostic factor and could effectively stratify patients with different risks. Patients with more high-expression CAMs had a higher risk of poor overall survival (OS) rate. The mortality risk for patients with elevation of >4 CAMs was 11 times of that in those without elevation of these 6 CAMs. Similar results were obtained when relapse-free survival (RFS) time was used during the analysis. Prognostic reliability of the six-gene model was validated using data of an independent cohort from the International Cancer Genome Consortium. In conclusion, a combination of CAM alterations contributed to progression and aggressiveness of EC. The six-gene signature was effective for predicting worse OS and RFS in patients with EC and could be complementary to the present clinical prognostic criteria.
Collapse
Affiliation(s)
- Xiangjun He
- Central Laboratory and Institute of Clinical Molecular Biology, Peking University People's Hospital, Beijing 100044, P.R. China
| | - Shu Lei
- Central Laboratory and Institute of Clinical Molecular Biology, Peking University People's Hospital, Beijing 100044, P.R. China.,Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing 100044, P.R. China
| | - Qi Zhang
- Central Laboratory and Institute of Clinical Molecular Biology, Peking University People's Hospital, Beijing 100044, P.R. China
| | - Liping Ma
- Central Laboratory and Institute of Clinical Molecular Biology, Peking University People's Hospital, Beijing 100044, P.R. China
| | - Na Li
- Central Laboratory and Institute of Clinical Molecular Biology, Peking University People's Hospital, Beijing 100044, P.R. China
| | - Jianliu Wang
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing 100044, P.R. China
| |
Collapse
|
45
|
Jiang Q, Xie Q, Hu C, Yang Z, Huang P, Shen H, Schachner M, Zhao W. Glioma malignancy is linked to interdependent and inverse AMOG and L1 adhesion molecule expression. BMC Cancer 2019; 19:911. [PMID: 31510944 PMCID: PMC6739972 DOI: 10.1186/s12885-019-6091-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 08/26/2019] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Gliomas account for the majority of primary human brain tumors and remain a challenging neoplasm for cure due to limited therapeutic options. Cell adhesion molecules play pivotal roles in the growth and progression of glial tumors. Roles of the adhesion molecules on glia (AMOG) and L1CAM (L1) in glioma cells have been shown to correlate with tumorigenesis: Increased expression of L1 and decreased expression of AMOG correlate with degree of malignancy. METHODS We evaluated the interdependence in expression of these molecules by investigating the role of AMOG in vitro via modulation of L1 expression and analyzing apoptosis and cell senescence of glioma cells. RESULTS Immunohistochemical staining of normal human cortical and glioma tissue microarrays demonstrated that AMOG expression was lower in human gliomas compared to normal tissue and is inversely correlated with the degree of malignancy. Moreover, reduction of AMOG expression in human glioblastoma cells elevated L1 expression, which is accompanied by decreased cell apoptosis as well as senescence. CONCLUSION AMOG and L1 interdependently regulate their expression levels not only in U-87 MG cells but also in U251 and SHG44 human glioma cell lines. The capacity of AMOG to reduce L1 expression suggests that methods for increasing AMOG expression may provide a therapeutic choice for the management of glial tumors with high expression of L1.
Collapse
Affiliation(s)
- Qiong Jiang
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong, 515041, People's Republic of China
| | - Qing Xie
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong, 515041, People's Republic of China
| | - Chengliang Hu
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong, 515041, People's Republic of China
| | - Zhai Yang
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong, 515041, People's Republic of China
| | - Peizhi Huang
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong, 515041, People's Republic of China
| | - Huifan Shen
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong, 515041, People's Republic of China
| | - Melitta Schachner
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong, 515041, People's Republic of China.
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ, 08854, USA.
| | - Weijiang Zhao
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong, 515041, People's Republic of China.
| |
Collapse
|
46
|
Spheroid glioblastoma culture conditions as antigen source for dendritic cell-based immunotherapy: spheroid proteins are survival-relevant targets but can impair immunogenic interferon γ production. Cytotherapy 2019; 21:643-658. [PMID: 30975602 DOI: 10.1016/j.jcyt.2019.03.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 01/04/2019] [Accepted: 03/02/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND Glioblastoma is the most aggressive type of brain cancer. Dendritic cell (DC)-based immunotherapy against glioblastoma depends on the effectiveness of loaded antigens. Sphere-inducing culture conditions are being studied by many as a potential antigen source. Here, we investigated two different in vitro conditions (spheroid culture versus adherent culture) in relation to DC immunotherapy: (1) We studied the specific spheroid-culture proteome and assessed the clinical importance of spheroid proteins. (2) We evaluated the immunogenicity of spheroid lysate - both compared to adherent conditions. METHODS We used seven spheroid culture systems, three of them patient-derived. Stemness-related markers were studied in those three via immunofluorescence. Spheroid-specific protein expression was measured via quantitative proteomics. The Cancer Genome Atlas (TCGA) survival data was used to investigate the clinical impact of spheroid proteins. Immunogenicity of spheroid versus adherent cell lysate was explored in autologous ELISPOT systems (DCs and T cells from the three patients). RESULTS (1) The differential proteome of spheroid versus adherent glioblastoma culture conditions could successfully be established. The top 10 identified spheroid-specific proteins were associated with significantly decreased overall survival (TCGA MIT/Harvard cohort; n = 350, P = 0.014). (2) In exploratory experiments, immunogenicity of spheroid lysate vis-á-vis interferon (IFN)γ production was lower than that of adherent cell lysate (IFNγ ELISPOT; P = 0.034). CONCLUSIONS Spheroid culture proteins seem to represent survival-relevant targets, supporting the use of spheroid culture conditions as an antigen source for DC immunotherapy. However, immunogenicity enhancement should be considered for future research. Transferability of our findings in terms of clinical impact and regarding different spheroid-generation techniques needs further validation.
Collapse
|
47
|
Pang JS, Li ZK, Lin P, Wang XD, Chen G, Yan HB, Li SH. The underlying molecular mechanism and potential drugs for treatment in papillary renal cell carcinoma: A study based on TCGA and Cmap datasets. Oncol Rep 2019; 41:2089-2102. [PMID: 30816528 PMCID: PMC6412146 DOI: 10.3892/or.2019.7014] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 02/05/2019] [Indexed: 12/23/2022] Open
Abstract
Papillary renal cell carcinoma (PRCC) accounts for 15–20% of all kidney neoplasms and continually attracts attention due to the increase in the incidents in which it occurs. The molecular mechanism of PRCC remains unclear and the efficacy of drugs that treat PRCC lacks sufficient evidence in clinical trials. Therefore, it is necessary to investigate the underlying mechanism in the development of PRCC and identify additional potential anti-PRCC drugs for its treatment. The differently expressed genes (DEGs) of PRCC were identified, followed by Gene Ontology and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses for functional annotation. Then, potential drugs for PRCC treatment were predicted by Connectivity Map (Cmap) based on DEGs. Furthermore, the latent function of query drugs in PRCC was explored by integrating drug-target, drug-pathway and drug-protein interactions. In total, 627 genes were screened as DEGs, and these DEGs were annotated using KEGG pathway analyses and were clearly associated with the complement and coagulation cascades, amongst others. Then, 60 candidate drugs, as predicted based on DEGs, were obtained from the Cmap database. Vorinostat was considered as the most promising drug for detailed discussion. Following protein-protein interaction (PPI) analysis and molecular docking, vorinostat was observed to interact with C3 and ANXN1 proteins, which are the upregulated hub genes and may serve as oncologic therapeutic targets in PRCC. Among the top 20 metabolic pathways, several significant pathways, such as complement and coagulation cascades and cell adhesion molecules, may greatly contribute to the development and progression of PRCC. Following the performance of the PPI network and molecular docking tests, vorinostat exhibited a considerable and promising application in PRCC treatment by targeting C3 and ANXN1.
Collapse
Affiliation(s)
- Jin-Shu Pang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Zhe-Kun Li
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Peng Lin
- Department of Medical Ultrasonics, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Xiao-Dong Wang
- Department of Medical Ultrasonics, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Gang Chen
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Hai-Biao Yan
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Sheng-Hua Li
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| |
Collapse
|
48
|
Sharifzad F, Ghavami S, Verdi J, Mardpour S, Mollapour Sisakht M, Azizi Z, Taghikhani A, Łos MJ, Fakharian E, Ebrahimi M, Hamidieh AA. Glioblastoma cancer stem cell biology: Potential theranostic targets. Drug Resist Updat 2019; 42:35-45. [PMID: 30877905 DOI: 10.1016/j.drup.2018.03.003] [Citation(s) in RCA: 111] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 02/28/2018] [Accepted: 03/16/2018] [Indexed: 12/21/2022]
Abstract
Glioblastoma multiforme (GBM) is among the most incurable cancers. GBMs survival rate has not markedly improved, despite new radical surgery protocols, the introduction of new anticancer drugs, new treatment protocols, and advances in radiation techniques. The low efficacy of therapy, and short interval between remission and recurrence, could be attributed to the resistance of a small fraction of tumorigenic cells to treatment. The existence and importance of cancer stem cells (CSCs) is perceived by some as controversial. Experimental evidences suggest that the presence of therapy-resistant glioblastoma stem cells (GSCs) could explain tumor recurrence and metastasis. Some scientists, including most of the authors of this review, believe that GSCs are the driving force behind GBM relapses, whereas others however, question the existence of GSCs. Evidence has accumulated indicating that non-tumorigenic cancer cells with high heterogeneity, could undergo reprogramming and become GSCs. Hence, targeting GSCs as the "root cells" initiating malignancy has been proposed to eradicate this devastating disease. Most standard treatments fail to completely eradicate GSCs, which can then cause the recurrence of the disease. To effectively target GSCs, a comprehensive understanding of the biology of GSCs as well as the mechanisms by which these cells survive during treatment and develop into new tumor, is urgently needed. Herein, we provide an overview of the molecular features of GSCs, and elaborate how to facilitate their detection and efficient targeting for therapeutic interventions. We also discuss GBM classifications based on the molecular stem cell subtypes with a focus on potential therapeutic approaches.
Collapse
Affiliation(s)
- Farzaneh Sharifzad
- Department of Applied Cell Sciences, Kashan University of Medical Sciences, Kashan, Iran; Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Saeid Ghavami
- Department of Human Anatomy & Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada; Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada; Research Institute of Oncology and Hematology, Cancer Care Manitoba, University of Manitoba, Winnipeg, Canada
| | - Javad Verdi
- Department of Applied Cell Sciences, Kashan University of Medical Sciences, Kashan, Iran; Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Soura Mardpour
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Zahra Azizi
- Heart Rhythm Program, Southlake Regional Health Centre, Toronto ON Canada
| | - Adeleh Taghikhani
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Immunology, Faculty of Medical Sciences, Tarbiat Modarres University, Tehran, Iran
| | - Marek J Łos
- Biotechnology Center, Silesian University of Technology in Gliwice, Poland
| | - Esmail Fakharian
- Department of Neurosurgery, Kashan University of Medical Sciences, Kashan, Iran
| | - Marzieh Ebrahimi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| | - Amir Ali Hamidieh
- Pediatric Stem Cell Transplant Department, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
49
|
Wang L, Bing T, Liu Y, Zhang N, Shen L, Liu X, Wang J, Shangguan D. Imaging of Neurite Network with an Anti-L1CAM Aptamer Generated by Neurite-SELEX. J Am Chem Soc 2018; 140:18066-18073. [PMID: 30485743 DOI: 10.1021/jacs.8b10783] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Neurite outgrowth is the critical step of nervous development. Molecular probes against neurites are essential for evaluation of the nervous system development, compound neurotoxicity, and drug efficacy on nerve regeneration. To obtain a neurite probe, we developed a neurite-SELEX strategy and generated a DNA aptamer, yly12, that strongly binds neurites. The molecular target of yly12 was identified to be neural cell adhesion molecule L1 (L1CAM), a surface antigen expressed in normal nervous system and various cancers. Here, yly12 was successfully applied to image the three-dimensional network of neurites between live cells, as well as the neurite fibers on normal brain tissue section. This aptamer was also found to have an inhibitory effect on neurite outgrowth between cells. Given the advantages of aptamers, yly12 hold great potential as a molecular tool in the field of neuroscientific research. The high efficiency of neurite-SELEX suggests that SELEX against a subcellular structure instead of the whole cells is more effective in obtaining the desired aptamers.
Collapse
Affiliation(s)
- Linlin Wang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences , Institute of Chemistry, Chinese Academy of Sciences , Beijing 100190 , China.,University of the Chinese Academy of Sciences , Beijing 100049 , China
| | - Tao Bing
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences , Institute of Chemistry, Chinese Academy of Sciences , Beijing 100190 , China.,University of the Chinese Academy of Sciences , Beijing 100049 , China
| | - Ying Liu
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences , Institute of Chemistry, Chinese Academy of Sciences , Beijing 100190 , China.,University of the Chinese Academy of Sciences , Beijing 100049 , China
| | - Nan Zhang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences , Institute of Chemistry, Chinese Academy of Sciences , Beijing 100190 , China.,University of the Chinese Academy of Sciences , Beijing 100049 , China
| | - Luyao Shen
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences , Institute of Chemistry, Chinese Academy of Sciences , Beijing 100190 , China.,University of the Chinese Academy of Sciences , Beijing 100049 , China
| | - Xiangjun Liu
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences , Institute of Chemistry, Chinese Academy of Sciences , Beijing 100190 , China.,University of the Chinese Academy of Sciences , Beijing 100049 , China
| | - Junyan Wang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences , Institute of Chemistry, Chinese Academy of Sciences , Beijing 100190 , China.,University of the Chinese Academy of Sciences , Beijing 100049 , China
| | - Dihua Shangguan
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences , Institute of Chemistry, Chinese Academy of Sciences , Beijing 100190 , China.,University of the Chinese Academy of Sciences , Beijing 100049 , China
| |
Collapse
|
50
|
A Subpopulation of Foxj1-Expressing, Nonmyelinating Schwann Cells of the Peripheral Nervous System Contribute to Schwann Cell Remyelination in the Central Nervous System. J Neurosci 2018; 38:9228-9239. [PMID: 30228229 PMCID: PMC6199410 DOI: 10.1523/jneurosci.0585-18.2018] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 07/09/2018] [Accepted: 07/31/2018] [Indexed: 12/18/2022] Open
Abstract
New myelin sheaths can be restored to demyelinated axons in a spontaneous regenerative process called remyelination. In general, new myelin sheaths are made by oligodendrocytes newly generated from a widespread population of adult CNS progenitors called oligodendrocyte progenitor cells (OPCs). New myelin in CNS remyelination in both experimental models and clinical diseases can also be generated by Schwann cells (SCs), the myelin-forming cells of the PNS. Fate-mapping studies have shown that SCs contributing to remyelination in the CNS are often derived from OPCs and appear not to be derived from myelinating SCs from the PNS. In this study, we address whether CNS remyelinating SCs can also be generated from PNS-derived cells other than myelinating SCs. Using a genetic fate-mapping approach, we have found that a subpopulation of nonmyelinating SCs identified by the expression of the transcription factor Foxj1 also contribute to CNS SC remyelination, as well as to remyelination in the PNS. We also find that the ependymal cells lining the central canal of the spinal cord, which also express Foxj1, do not generate cells that contribute to CNS remyelination. These findings therefore identify a previously unrecognized population of PNS glia that can participate in the regeneration of new myelin sheaths following CNS demyelination.SIGNIFICANCE STATEMENT Remyelination failure in chronic demyelinating diseases such as multiple sclerosis drives the current quest for developing means by which remyelination in CNS can be enhanced therapeutically. Critical to this endeavor is the need to understand the mechanisms of remyelination, including the nature and identity of the cells capable of generating new myelin sheath-forming cells. Here, we report a previously unrecognized subpopulation of nonmyelinating Schwann cells (SCs) in the PNS, identified by the expression of the transcription factor Foxj1, which can give rise to SCs that are capable of remyelinating both PNS and CNS axons. These cells therefore represent a new cellular target for myelin regenerative strategies for the treatment of CNS disorders characterized by persistent demyelination.
Collapse
|