1
|
Miyagishima KJ, Qiao F, Stasheff SF, Nadal-Nicolás FM. Visual Deficits and Diagnostic and Therapeutic Strategies for Neurofibromatosis Type 1: Bridging Science and Patient-Centered Care. Vision (Basel) 2024; 8:31. [PMID: 38804352 PMCID: PMC11130890 DOI: 10.3390/vision8020031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 05/03/2024] [Accepted: 05/04/2024] [Indexed: 05/29/2024] Open
Abstract
Neurofibromatosis type 1 (NF1) is an inherited autosomal dominant disorder primarily affecting children and adolescents characterized by multisystemic clinical manifestations. Mutations in neurofibromin, the protein encoded by the Nf1 tumor suppressor gene, result in dysregulation of the RAS/MAPK pathway leading to uncontrolled cell growth and migration. Neurofibromin is highly expressed in several cell lineages including melanocytes, glial cells, neurons, and Schwann cells. Individuals with NF1 possess a genetic predisposition to central nervous system neoplasms, particularly gliomas affecting the visual pathway, known as optic pathway gliomas (OPGs). While OPGs are typically asymptomatic and benign, they can induce visual impairment in some patients. This review provides insight into the spectrum and visual outcomes of NF1, current diagnostic techniques and therapeutic interventions, and explores the influence of NF1-OPGS on visual abnormalities. We focus on recent advancements in preclinical animal models to elucidate the underlying mechanisms of NF1 pathology and therapies targeting NF1-OPGs. Overall, our review highlights the involvement of retinal ganglion cell dysfunction and degeneration in NF1 disease, and the need for further research to transform scientific laboratory discoveries to improved patient outcomes.
Collapse
Affiliation(s)
- Kiyoharu J. Miyagishima
- Retinal Neurophysiology Section, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA; (K.J.M.); (F.Q.); (S.F.S.)
| | - Fengyu Qiao
- Retinal Neurophysiology Section, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA; (K.J.M.); (F.Q.); (S.F.S.)
| | - Steven F. Stasheff
- Retinal Neurophysiology Section, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA; (K.J.M.); (F.Q.); (S.F.S.)
- Center for Neuroscience and Behavioral Medicine, Gilbert Neurofibromatosis Institute, Children’s National Health System, Washington, DC 20010, USA
- Neurology Department, George Washington University School of Medicine, Washington, DC 20037, USA
| | - Francisco M. Nadal-Nicolás
- Retinal Neurophysiology Section, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA; (K.J.M.); (F.Q.); (S.F.S.)
| |
Collapse
|
2
|
Atsoniou K, Giannopoulou E, Georganta EM, Skoulakis EMC. Drosophila Contributions towards Understanding Neurofibromatosis 1. Cells 2024; 13:721. [PMID: 38667335 PMCID: PMC11048932 DOI: 10.3390/cells13080721] [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: 03/15/2024] [Revised: 04/17/2024] [Accepted: 04/19/2024] [Indexed: 04/28/2024] Open
Abstract
Neurofibromatosis 1 (NF1) is a multisymptomatic disorder with highly variable presentations, which include short stature, susceptibility to formation of the characteristic benign tumors known as neurofibromas, intense freckling and skin discoloration, and cognitive deficits, which characterize most children with the condition. Attention deficits and Autism Spectrum manifestations augment the compromised learning presented by most patients, leading to behavioral problems and school failure, while fragmented sleep contributes to chronic fatigue and poor quality of life. Neurofibromin (Nf1) is present ubiquitously during human development and postnatally in most neuronal, oligodendrocyte, and Schwann cells. Evidence largely from animal models including Drosophila suggests that the symptomatic variability may reflect distinct cell-type-specific functions of the protein, which emerge upon its loss, or mutations affecting the different functional domains of the protein. This review summarizes the contributions of Drosophila in modeling multiple NF1 manifestations, addressing hypotheses regarding the cell-type-specific functions of the protein and exploring the molecular pathways affected upon loss of the highly conserved fly homolog dNf1. Collectively, work in this model not only has efficiently and expediently modelled multiple aspects of the condition and increased understanding of its behavioral manifestations, but also has led to pharmaceutical strategies towards their amelioration.
Collapse
Affiliation(s)
- Kalliopi Atsoniou
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Center “Alexander Fleming”, 16672 Athens, Greece; (K.A.); (E.G.)
- Laboratory of Experimental Physiology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Eleni Giannopoulou
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Center “Alexander Fleming”, 16672 Athens, Greece; (K.A.); (E.G.)
| | - Eirini-Maria Georganta
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Center “Alexander Fleming”, 16672 Athens, Greece; (K.A.); (E.G.)
| | - Efthimios M. C. Skoulakis
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Center “Alexander Fleming”, 16672 Athens, Greece; (K.A.); (E.G.)
| |
Collapse
|
3
|
Creus‐Bachiller E, Fernández‐Rodríguez J, Magallón‐Lorenz M, Ortega‐Bertran S, Navas‐Rutete S, Romagosa C, Silva TM, Pané M, Estival A, Perez Sidelnikova D, Morell M, Mazuelas H, Carrió M, Lausová T, Reuss D, Gel B, Villanueva A, Serra E, Lázaro C. Expanding a precision medicine platform for malignant peripheral nerve sheath tumors: New patient-derived orthotopic xenografts, cell lines and tumor entities. Mol Oncol 2024; 18:895-917. [PMID: 37798904 PMCID: PMC10994238 DOI: 10.1002/1878-0261.13534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 08/07/2023] [Accepted: 10/04/2023] [Indexed: 10/07/2023] Open
Abstract
Malignant peripheral nerve sheath tumors (MPNSTs) are aggressive soft-tissue sarcomas with a poor survival rate, presenting either sporadically or in the context of neurofibromatosis type 1 (NF1). The histological diagnosis of MPNSTs can be challenging, with different tumors exhibiting great histological and marker expression overlap. This heterogeneity could be partly responsible for the observed disparity in treatment response due to the inherent diversity of the preclinical models used. For several years, our group has been generating a large patient-derived orthotopic xenograft (PDOX) MPNST platform for identifying new precision medicine treatments. Herein, we describe the expansion of this platform using six primary tumors clinically diagnosed as MPNSTs, from which we obtained six additional PDOX mouse models and three cell lines, thus generating three pairs of in vitro-in vivo models. We extensively characterized these tumors and derived preclinical models, including genomic, epigenomic, and histological analyses. Tumors were reclassified after these analyses: three remained as MPNSTs (two being classic MPNSTs), one was a melanoma, another was a neurotrophic tyrosine receptor kinase (NTRK)-rearranged spindle cell neoplasm, and, finally, the last was an unclassifiable tumor bearing neurofibromin-2 (NF2) inactivation, a neuroblastoma RAS viral oncogene homolog (NRAS) oncogenic mutation, and a SWI/SNF-related matrix-associated actin-dependent regulator of chromatin (SMARCA4) heterozygous truncated variant. New cell lines and PDOXs faithfully recapitulated histology, marker expression, and genomic characteristics of the primary tumors. The diversity in tumor identity and their specific associated genomic alterations impacted treatment responses obtained when we used the new cell lines for testing compounds against known altered pathways in MPNSTs. In summary, we present here an extension of our MPNST precision medicine platform, with new PDOXs and cell lines, including tumor entities confounded as MPNSTs in a real clinical scenario. This platform may constitute a useful tool for obtaining correct preclinical information to guide MPNST clinical trials.
Collapse
Affiliation(s)
- Edgar Creus‐Bachiller
- Hereditary Cancer ProgramCatalan Institute of Oncology, ICO‐IDIBELL, Hospitalet de LlobregatBarcelonaSpain
- Program in Molecular Mechanisms and Experimental Therapy in Oncology (Oncobell), IDIBELL, Hospitalet de LlobregatBarcelonaSpain
| | - Juana Fernández‐Rodríguez
- Hereditary Cancer ProgramCatalan Institute of Oncology, ICO‐IDIBELL, Hospitalet de LlobregatBarcelonaSpain
- Program in Molecular Mechanisms and Experimental Therapy in Oncology (Oncobell), IDIBELL, Hospitalet de LlobregatBarcelonaSpain
- Mouse Lab, IDIBELL, Hospitalet de LlobregatBarcelonaSpain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC)MadridSpain
| | | | - Sara Ortega‐Bertran
- Hereditary Cancer ProgramCatalan Institute of Oncology, ICO‐IDIBELL, Hospitalet de LlobregatBarcelonaSpain
- Program in Molecular Mechanisms and Experimental Therapy in Oncology (Oncobell), IDIBELL, Hospitalet de LlobregatBarcelonaSpain
| | - Susana Navas‐Rutete
- Hereditary Cancer ProgramCatalan Institute of Oncology, ICO‐IDIBELL, Hospitalet de LlobregatBarcelonaSpain
| | | | - Tulio M. Silva
- Department of PathologyHospital Vall d'HebronBarcelonaSpain
| | - Maria Pané
- Department of PathologyHUB‐IDIBELL, L'Hospitalet de LlobregatBarcelonaSpain
| | - Anna Estival
- Department of Medical OncologyCatalan Institute of OncologyBarcelonaSpain
| | | | - Mireia Morell
- Hereditary Cancer ProgramCatalan Institute of Oncology, ICO‐IDIBELL, Hospitalet de LlobregatBarcelonaSpain
- Program in Molecular Mechanisms and Experimental Therapy in Oncology (Oncobell), IDIBELL, Hospitalet de LlobregatBarcelonaSpain
- Mouse Lab, IDIBELL, Hospitalet de LlobregatBarcelonaSpain
| | - Helena Mazuelas
- Hereditary Cancer Group, Germans Trias i Pujol Research Institute (IGTP)BarcelonaSpain
| | - Meritxell Carrió
- Hereditary Cancer Group, Germans Trias i Pujol Research Institute (IGTP)BarcelonaSpain
| | - Tereza Lausová
- Department of NeuropathologyInstitute of Pathology, Heidelberg University HospitalHeidelbergGermany
- Clinical Cooperation Unit NeuropathologyGerman Cancer Research Center (DKFZ), German Consortium for Translational Cancer Research (DKTK)HeidelbergGermany
| | - David Reuss
- Department of NeuropathologyInstitute of Pathology, Heidelberg University HospitalHeidelbergGermany
- Clinical Cooperation Unit NeuropathologyGerman Cancer Research Center (DKFZ), German Consortium for Translational Cancer Research (DKTK)HeidelbergGermany
| | - Bernat Gel
- Hereditary Cancer Group, Germans Trias i Pujol Research Institute (IGTP)BarcelonaSpain
| | - Alberto Villanueva
- Program in Molecular Mechanisms and Experimental Therapy in Oncology (Oncobell), IDIBELL, Hospitalet de LlobregatBarcelonaSpain
- Procure ProgramCatalan Institute of OncologyBarcelonaSpain
| | - Eduard Serra
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC)MadridSpain
- Hereditary Cancer Group, Germans Trias i Pujol Research Institute (IGTP)BarcelonaSpain
| | - Conxi Lázaro
- Hereditary Cancer ProgramCatalan Institute of Oncology, ICO‐IDIBELL, Hospitalet de LlobregatBarcelonaSpain
- Program in Molecular Mechanisms and Experimental Therapy in Oncology (Oncobell), IDIBELL, Hospitalet de LlobregatBarcelonaSpain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC)MadridSpain
| |
Collapse
|
4
|
Pu F, Guo H, Shi D, Chen F, Peng Y, Huang X, Liu J, Zhang Z, Shao Z. The generation and use of animal models of osteosarcoma in cancer research. Genes Dis 2024; 11:664-674. [PMID: 37692517 PMCID: PMC10491873 DOI: 10.1016/j.gendis.2022.12.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 12/16/2022] [Indexed: 09/12/2023] Open
Abstract
Osteosarcoma is the most common malignant bone tumor affecting children and adolescents. Currently, the most common treatment is surgery combined with neoadjuvant chemotherapy. Although the survival rate of patients with osteosarcoma has improved in recent years, it remains poor when the tumor(s) progress and distant metastases develop. Therefore, better animal models that more accurately replicate the natural progression of the disease are needed to develop improved prognostic and diagnostic markers, as well as targeted therapies for both primary and metastatic osteosarcoma. The present review described animal models currently being used in research investigating osteosarcoma, and their characteristics, advantages, and disadvantages. These models may help elucidate the pathogenic mechanism(s) of osteosarcoma and provide evidence to support and develop clinical treatment strategies.
Collapse
Affiliation(s)
- Feifei Pu
- Department of Orthopedics, Wuhan Hospital of Traditional Chinese and Western Medicine (Wuhan No.1 Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Haoyu Guo
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Deyao Shi
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Fengxia Chen
- Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei 430071, China
- Hubei Cancer Clinical Study Center, Zhongnan Hospital, Wuhan University, Wuhan, Hubei 430071, China
| | - Yizhong Peng
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Xin Huang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Jianxiang Liu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Zhicai Zhang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Zengwu Shao
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| |
Collapse
|
5
|
Mei C, Zhang X, Zhi Y, Liang Z, Xu H, Liu Z, Liu Y, Lyu Y, Wang H. Isorhamnetin Regulates Programmed Death Ligand-1 Expression by Suppressing the EGFR-STAT3 Signaling Pathway in Canine Mammary Tumors. Int J Mol Sci 2024; 25:670. [PMID: 38203840 PMCID: PMC10779303 DOI: 10.3390/ijms25010670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 12/12/2023] [Accepted: 12/19/2023] [Indexed: 01/12/2024] Open
Abstract
Programmed death ligand-1 (PD-L1) is highly expressed in a variety of cancer cells and suggests a poorer prognosis for patients. The natural compound isorhamnetin (ISO) shows promise in treating cancers and causing damage to canine mammary tumor (CMT) cells. We investigated the mechanism of ISO in reducing PD-L1 expression in CMT cells. Clustered, regularly interspaced short palindromic repeat-associated protein 9 (CRISPR/Cas9) was used to mediate CD274 knockout in U27 cells. Then, monoclonal cells were screened and cultured. Nucleotide sequencing and expression of PD-L1 were detected. Additionally, we examined cell migration, invasion, and damage. Immunofluorescent staining of PD-L1 was examined in U27 cells. The signaling pathways were measured by Western blotting. Murine xenotransplantation models and murine immunocompetent allograft mammary tumor models were established to evaluate the effect of ISO therapy. Expression of Ki-67, caspase3, and PD-L1 were analyzed by immunohistochemistry. A pull-down assay was used to explore which proteins could bind to ISO. Canine EGFR protein was purified and used to detect whether it directly binds to ISO using a surface plasmon resonance assay. ISO inhibited the EGFR-STAT3-PD-L1 signaling pathway and blocked cancer growth, significantly increasing the survival rate of healthy cells. The cell membrane receptor EGFR was identified as a direct target of ISO. ISO could be exploited as an antineoplastic treatment of CMT by targeting EGFR to suppress PD-L1 expression.
Collapse
Affiliation(s)
- Chen Mei
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, No. 11 Shuguanghuayuan Middle Road, Haidian District, Beijing 100097, China; (C.M.); (X.Z.); (Y.Z.); (Z.L.); (H.X.); (Z.L.); (Y.L.)
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Haidian District, Beijing 100193, China
| | - Xue Zhang
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, No. 11 Shuguanghuayuan Middle Road, Haidian District, Beijing 100097, China; (C.M.); (X.Z.); (Y.Z.); (Z.L.); (H.X.); (Z.L.); (Y.L.)
| | - Yan Zhi
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, No. 11 Shuguanghuayuan Middle Road, Haidian District, Beijing 100097, China; (C.M.); (X.Z.); (Y.Z.); (Z.L.); (H.X.); (Z.L.); (Y.L.)
| | - Zhixuan Liang
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, No. 11 Shuguanghuayuan Middle Road, Haidian District, Beijing 100097, China; (C.M.); (X.Z.); (Y.Z.); (Z.L.); (H.X.); (Z.L.); (Y.L.)
| | - Haojun Xu
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, No. 11 Shuguanghuayuan Middle Road, Haidian District, Beijing 100097, China; (C.M.); (X.Z.); (Y.Z.); (Z.L.); (H.X.); (Z.L.); (Y.L.)
| | - Zhenyi Liu
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, No. 11 Shuguanghuayuan Middle Road, Haidian District, Beijing 100097, China; (C.M.); (X.Z.); (Y.Z.); (Z.L.); (H.X.); (Z.L.); (Y.L.)
| | - Ying Liu
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, No. 11 Shuguanghuayuan Middle Road, Haidian District, Beijing 100097, China; (C.M.); (X.Z.); (Y.Z.); (Z.L.); (H.X.); (Z.L.); (Y.L.)
| | - Yanli Lyu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Haidian District, Beijing 100193, China
| | - Hongjun Wang
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, No. 11 Shuguanghuayuan Middle Road, Haidian District, Beijing 100097, China; (C.M.); (X.Z.); (Y.Z.); (Z.L.); (H.X.); (Z.L.); (Y.L.)
| |
Collapse
|
6
|
Li S, Lu Z, Wu S, Chu T, Li B, Qi F, Zhao Y, Nie G. The dynamic role of platelets in cancer progression and their therapeutic implications. Nat Rev Cancer 2024; 24:72-87. [PMID: 38040850 DOI: 10.1038/s41568-023-00639-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/13/2023] [Indexed: 12/03/2023]
Abstract
Systemic antiplatelet treatment represents a promising option to improve the therapeutic outcomes and therapeutic efficacy of chemotherapy and immunotherapy due to the critical contribution of platelets to tumour progression. However, until recently, targeting platelets as a cancer therapeutic has been hampered by the elevated risk of haemorrhagic and thrombocytopenic (low platelet count) complications owing to the lack of specificity for tumour-associated platelets. Recent work has advanced our understanding of the molecular mechanisms responsible for the contribution of platelets to tumour progression and metastasis. This has led to the identification of the biological changes in platelets in the presence of tumours, the complex interactions between platelets and tumour cells during tumour progression, and the effects of platelets on antitumour therapeutic response. In this Review, we present a detailed picture of the dynamic roles of platelets in tumour development and progression as well as their use in diagnosis, prognosis and monitoring response to therapy. We also provide our view on how to overcome challenges faced by the development of precise antiplatelet strategies for safe and efficient clinical cancer therapy.
Collapse
Affiliation(s)
- Suping Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China.
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, China.
| | - Zefang Lu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Suying Wu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Tianjiao Chu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- College of Pharmaceutical Science, Jilin University, Changchun, China
| | - Bozhao Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Feilong Qi
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, China
- Department of Chemistry, Tsinghua University, Beijing, China
| | - Yuliang Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China.
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
7
|
Brockman QR, Scherer A, McGivney GR, Gutierrez WR, Rytlewski J, Sheehan A, Warrier A, Laverty EA, Roughton G, Carnevale NC, Knepper-Adrian V, Dodd RD. Discrepancies in indel software resolution with somatic CRISPR/Cas9 tumorigenesis models. Sci Rep 2023; 13:14798. [PMID: 37684258 PMCID: PMC10491828 DOI: 10.1038/s41598-023-41109-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 08/22/2023] [Indexed: 09/10/2023] Open
Abstract
CRISPR/Cas9 gene editing has evolved from a simple laboratory tool to a powerful method of in vivo genomic engineering. As the applications of CRISPR/Cas9 technology have grown, the need to characterize the breadth and depth of indels generated by editing has expanded. Traditionally, investigators use one of several publicly-available platforms to determine CRISPR/Cas9-induced indels in an edited sample. However, to our knowledge, there has not been a cross-platform comparison of available indel analysis software in samples generated from somatic in vivo mouse models. Our group has pioneered using CRISPR/Cas9 to generate somatic primary mouse models of malignant peripheral nerve sheath tumors (MPNSTs) through genetic editing of Nf1. Here, we used sequencing data from the in vivo editing of the Nf1 gene in our CRISPR/Cas9 tumorigenesis model to directly compare results across four different software platforms. By analyzing the same genetic target across a wide panel of cell lines with the same sequence file, we are able to draw systematic conclusions about the differences in these software programs for analysis of in vivo-generated indels. Surprisingly, we report high variability in the reported number, size, and frequency of indels across each software platform. These data highlight the importance of selecting indel analysis platforms specific to the context that the gene editing approach is being applied. Taken together, this analysis shows that different software platforms can report widely divergent indel data from the same sample, particularly if larger indels are present, which are common in somatic, in vivo CRISPR/Cas9 tumor models.
Collapse
Affiliation(s)
- Qierra R Brockman
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, 375 Newton Rd, 5206 MERF, Iowa City, IA, 52246, USA
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA
| | - Amanda Scherer
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, 375 Newton Rd, 5206 MERF, Iowa City, IA, 52246, USA
| | - Gavin R McGivney
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, 375 Newton Rd, 5206 MERF, Iowa City, IA, 52246, USA
- Cancer Biology Training Program, University of Iowa, Iowa City, IA, USA
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA, USA
| | - Wade R Gutierrez
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, 375 Newton Rd, 5206 MERF, Iowa City, IA, 52246, USA
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA
- Cancer Biology Training Program, University of Iowa, Iowa City, IA, USA
- Medical Scientist Training Program, University of Iowa, Iowa City, IA, USA
| | - Jeffrey Rytlewski
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, 375 Newton Rd, 5206 MERF, Iowa City, IA, 52246, USA
| | - Alexa Sheehan
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, 375 Newton Rd, 5206 MERF, Iowa City, IA, 52246, USA
| | - Akshaya Warrier
- Cancer Biology Training Program, University of Iowa, Iowa City, IA, USA
- Medical Scientist Training Program, University of Iowa, Iowa City, IA, USA
| | - Emily A Laverty
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, 375 Newton Rd, 5206 MERF, Iowa City, IA, 52246, USA
| | - Grace Roughton
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, 375 Newton Rd, 5206 MERF, Iowa City, IA, 52246, USA
| | - Nina C Carnevale
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, 375 Newton Rd, 5206 MERF, Iowa City, IA, 52246, USA
| | - Vickie Knepper-Adrian
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, 375 Newton Rd, 5206 MERF, Iowa City, IA, 52246, USA
| | - Rebecca D Dodd
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, 375 Newton Rd, 5206 MERF, Iowa City, IA, 52246, USA.
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA.
- Cancer Biology Training Program, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
8
|
Knoernschild K, Johnson HJ, Schroeder KE, Swier VJ, White KA, Sato TS, Rogers CS, Weimer JM, Sieren JC. Magnetic resonance brain volumetry biomarkers of CLN2 Batten disease identified with miniswine model. Sci Rep 2023; 13:5146. [PMID: 36991106 PMCID: PMC10060411 DOI: 10.1038/s41598-023-32071-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 03/22/2023] [Indexed: 03/31/2023] Open
Abstract
Late-infantile neuronal ceroid lipofuscinosis type 2 (CLN2) disease (Batten disease) is a rare pediatric disease, with symptom development leading to clinical diagnosis. Early diagnosis and effective tracking of disease progression are required for treatment. We hypothesize that brain volumetry is valuable in identifying CLN2 disease at an early stage and tracking disease progression in a genetically modified miniswine model. CLN2R208X/R208X miniswine and wild type controls were evaluated at 12- and 17-months of age, correlating to early and late stages of disease progression. Magnetic resonance imaging (MRI) T1- and T2-weighted data were acquired. Total intercranial, gray matter, cerebrospinal fluid, white matter, caudate, putamen, and ventricle volumes were calculated and expressed as proportions of the intracranial volume. The brain regions were compared between timepoints and cohorts using Gardner-Altman plots, mean differences, and confidence intervals. At an early stage of disease, the total intracranial volume (- 9.06 cm3), gray matter (- 4.37% 95 CI - 7.41; - 1.83), caudate (- 0.16%, 95 CI - 0.24; - 0.08) and putamen (- 0.11% 95 CI - 0.23; - 0.02) were all notably smaller in CLN2R208X/R208X miniswines versus WT, while cerebrospinal fluid was larger (+ 3.42%, 95 CI 2.54; 6.18). As the disease progressed to a later stage, the difference between the gray matter (- 8.27%, 95 CI - 10.1; - 5.56) and cerebrospinal fluid (+ 6.88%, 95 CI 4.31; 8.51) continued to become more pronounced, while others remained stable. MRI brain volumetry in this miniswine model of CLN2 disease is sensitive to early disease detection and longitudinal change monitoring, providing a valuable tool for pre-clinical treatment development and evaluation.
Collapse
Affiliation(s)
- Kevin Knoernschild
- Department of Radiology, University of Iowa, 200 Hawkins Drive cc704 GH, Iowa City, IA, 52242, USA
- Department of Biomedical Engineering, University of Iowa, Iowa City, IA, USA
| | - Hans J Johnson
- Department of Biomedical Engineering, University of Iowa, Iowa City, IA, USA
- Department of Electrical and Computer Engineering, University of Iowa, Iowa City, IA, USA
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
| | - Kimberly E Schroeder
- Department of Radiology, University of Iowa, 200 Hawkins Drive cc704 GH, Iowa City, IA, 52242, USA
| | - Vicki J Swier
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, USA
| | - Katherine A White
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, USA
| | - Takashi S Sato
- Department of Radiology, University of Iowa, 200 Hawkins Drive cc704 GH, Iowa City, IA, 52242, USA
| | | | - Jill M Weimer
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, USA
| | - Jessica C Sieren
- Department of Radiology, University of Iowa, 200 Hawkins Drive cc704 GH, Iowa City, IA, 52242, USA.
- Department of Biomedical Engineering, University of Iowa, Iowa City, IA, USA.
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
9
|
Tekavec K, Švara T, Knific T, Mlakar J, Gombač M, Cantile C. Loss of H3K27me3 expression in canine nerve sheath tumors. Front Vet Sci 2022; 9:921720. [PMID: 35968018 PMCID: PMC9372589 DOI: 10.3389/fvets.2022.921720] [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: 04/16/2022] [Accepted: 07/13/2022] [Indexed: 11/13/2022] Open
Abstract
Nerve sheath tumors (NSTs) are characterized by neoplastic proliferation of Schwann cells, perineurial cells, endoneurial and/or epineurial fibroblasts. Diagnosis of NST is often challenging, particularly in distinguishing malignant NST (MNST) from other soft tissue sarcomas, or sometimes between low-grade MNST and benign NST. Recent studies in human pathology have demonstrated loss of trimethylation at lysine 27 of histone 3 (H3K27me3) in a subset of MNSTs using immunohistochemistry. Loss of H3K27me3 expression is rare in other high-grade sarcomas and also appears to be useful in distinguishing benign and low-grade MNSTs from high-grade subsets. In our retrospective study, we performed H3K27me3 immunohistochemistry in 68 canine tumors previously diagnosed as NST. We detected loss of H3K27me3 expression in 25% (n = 17) of all canine NST, including one neurofibroma, whereas 49% (n = 33) of tumors had mosaic loss of expression and 26% (n = 18) retained expression. No statistically significant differences were found between H3K27me3 expression, histopathological features of tumors, and their immunoreactivity for Sox10, claudin-1, GFAP, and Ki67. Because the classification of canine NST is not yet fully established and its correlation with the prognosis and clinical course of the disease is lacking, prospective studies with possible genetic analyses are needed to assess the true diagnostic value of H3K27me3 loss in canine NST.
Collapse
Affiliation(s)
- Kristina Tekavec
- Department of Veterinary Science, University of Pisa, Pisa, Italy
- Veterinary Faculty, Institute of Pathology, Wild Animals, Fish and Bees, University of Ljubljana, Ljubljana, Slovenia
- *Correspondence: Kristina Tekavec
| | - Tanja Švara
- Veterinary Faculty, Institute of Pathology, Wild Animals, Fish and Bees, University of Ljubljana, Ljubljana, Slovenia
| | - Tanja Knific
- Veterinary Faculty, Institute of Food Safety, Feed and Environment, University of Ljubljana, Ljubljana, Slovenia
| | - Jernej Mlakar
- Faculty of Medicine, Institute of Pathology, University of Ljubljana, Ljubljana, Slovenia
| | - Mitja Gombač
- Veterinary Faculty, Institute of Pathology, Wild Animals, Fish and Bees, University of Ljubljana, Ljubljana, Slovenia
| | - Carlo Cantile
- Department of Veterinary Science, University of Pisa, Pisa, Italy
| |
Collapse
|
10
|
Marsh O, Shimizu N, Mason SL, Uriarte A. Case Report: A Novel Lateral Approach to the C7, C8, and T1 Intervertebral Foramina for Resection of Malignant Peripheral Nerve Sheath Neoplasia, Followed by Adjunctive Radiotherapy, in Three Dogs. Front Vet Sci 2022; 9:869082. [PMID: 35720837 PMCID: PMC9204227 DOI: 10.3389/fvets.2022.869082] [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: 02/03/2022] [Accepted: 05/06/2022] [Indexed: 11/17/2022] Open
Abstract
This case report describes the diagnosis, management and outcome of three dogs with peripheral nerve sheath tumors (PNSTs) involving the brachial plexus, C7 (case 1), C8 (case 2), and C8 and T1 (case 3) spinal nerves and nerve roots with intrathoracic invasion. Surgical resection required thoracic limb amputation and removal of the first rib, facilitating a novel lateral approach to the spinal nerves and foramina in all cases. This was followed by hemilaminectomy and rhizotomy in cases 1 and 2. Adjunctive radiotherapy was then performed in all dogs. All three dogs regained a good quality of life in the short-term following surgery. Two were euthanased after 3 and 10 months, following detection of a pulmonary mass in one case and multiple thoracic and abdominal masses in the other. The third dog was alive and well at the time of writing (7 months post-surgery). This surgical approach facilitated good access and allowed gross neoplastic tissue to be resected. The ease of surgical access was dependent, to a degree, on the size of the patient. This surgical approach can be considered in cases of PNSTs involving the caudal cervical or cranial thoracic spinal nerves and nerve roots. Adjunctive radiotherapy should be considered as part of a multi-modal approach to these challenging tumors due to the difficulty of achieving clean margins, particularly proximally, even with optimal surgical access.
Collapse
Affiliation(s)
- Oliver Marsh
- Linnaeus Veterinary Limited, Neurology and Neurosurgery Service, Southfields Veterinary Specialists, Essex, United Kingdom
| | - Naomi Shimizu
- Linnaeus Veterinary Limited, Orthopaedic and Soft Tissue Surgery Service, Southfields Veterinary Specialists, Essex, United Kingdom
| | - Sarah L. Mason
- Linnaeus Veterinary Limited, Oncology Service, Southfields Veterinary Specialists, Essex, United Kingdom
| | - Ane Uriarte
- Linnaeus Veterinary Limited, Neurology and Neurosurgery Service, Southfields Veterinary Specialists, Essex, United Kingdom
| |
Collapse
|
11
|
Mu X, Zhang HY, Shen YH, Yang HY. Familial left cervical neurofibromatosis 1 with scoliosis: A case report. World J Clin Cases 2021; 9:8839-8845. [PMID: 34734064 PMCID: PMC8546810 DOI: 10.12998/wjcc.v9.i29.8839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/18/2021] [Accepted: 08/18/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Neurofibromatosis type 1 (NF1) is an inherited autosomal dominant disorder affecting many parts of the body with café au lait spots, skeletal deformity, and scoliosis. A familial case of NF1 with scoliosis and a painless mass had not yet been reported.
CASE SUMMARY We describe the case of a 15-year-old male patient with a painless lump on the left side of his neck for 10 years and scoliosis. His right shoulder was about 5 cm lower than the left, the left side of his face was deformed, and the left submandibular skin was relaxed. The folding and drooping were obvious and movement was poor. Computed tomography revealed the involvement of the neck, upper chest wall, and surrounding left shoulder, accompanied by bone changes and scoliosis. Histological evaluation showed subepidermal pale blue mucoid degeneration, fibrous fusiform cells in the dermis in a fascicular, woven arrangement. His mother had the same medical history. The diagnosis was neurofibromatosis of the left neck. Various parts of the tumor tissue were serially resected during several visits. Eight months after surgery, there was a slight tendency to regrow.
CONCLUSION This case of slow-progressing NF1 highlights the importance of early diagnosis and treatment to reduce its impact on the patient’s growth and development.
Collapse
Affiliation(s)
- Xia Mu
- School of Stomatology, Zunyi Medical University, Zunyi 563000, Guizhou Province, China
- Department of Stomatology, Peking University Shenzhen Hospital, Shenzhen 518036, Guangdong Province, China
| | - Han-Yu Zhang
- Department of Oral and Maxillofacial Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, Guangdong Province, China
| | - Yue-Hong Shen
- Department of Stomatology, Peking University Shenzhen Hospital, Shenzhen 518036, Guangdong Province, China
| | - Hong-Yu Yang
- School of Stomatology, Zunyi Medical University, Zunyi 563000, Guizhou Province, China
- Department of Stomatology, Peking University Shenzhen Hospital, Shenzhen 518036, Guangdong Province, China
| |
Collapse
|