scholarly journals IMMU-28. DEFINING MOLECULAR MECHANISMS OF RESISTANCE TO GLIOBLASTOMA (GBM) IMMUNITY USING A NOVEL CRISPR/CAS9 IN VIVO LOSS-OF-FUNCTION SCREENING PLATFORM

2017 ◽  
Vol 19 (suppl_6) ◽  
pp. vi118-vi118
Author(s):  
Martha R Neagu ◽  
Maria Carmela Speranza ◽  
Robert T Manguso ◽  
Sean E Lawler ◽  
Gordon J Freeman ◽  
...  
2017 ◽  
Vol 2 (1) ◽  
Author(s):  
Dalia Martinez-Marin ◽  
Courtney Jarvis ◽  
Thomas Nelius ◽  
Stéphanie Filleur

Abstract Macrophages have been recognized as the main inflammatory component of the tumor microenvironment. Although often considered as beneficial for tumor growth and disease progression, tumor-associated macrophages have also been shown to be detrimental to the tumor depending on the tumor microenvironment. Therefore, understanding the molecular interactions between macrophages and tumor cells in relation to macrophages functional activities such as phagocytosis is critical for a better comprehension of their tumor-modulating action. Still, the characterization of these molecular mechanisms in vivo remains complicated due to the extraordinary complexity of the tumor microenvironment and the broad range of tumor-associated macrophage functions. Thus, there is an increasing demand for in vitro methodologies to study the role of cell–cell interactions in the tumor microenvironment. In the present study, we have developed live co-cultures of macrophages and human prostate tumor cells to assess the phagocytic activity of macrophages using a combination of Confocal and Nomarski Microscopy. Using this model, we have emphasized that this is a sensitive, measurable, and highly reproducible functional assay. We have also highlighted that this assay can be applied to multiple cancer cell types and used as a selection tool for a variety of different types of phagocytosis agonists. Finally, combining with other studies such as gain/loss of function or signaling studies remains possible. A better understanding of the interactions between tumor cells and macrophages may lead to the identification of new therapeutic targets against cancer.


Thorax ◽  
2020 ◽  
Vol 75 (6) ◽  
pp. 486-493 ◽  
Author(s):  
Ling Chu ◽  
Yongfeng Luo ◽  
Hui Chen ◽  
Qing Miao ◽  
Larry Wang ◽  
...  

BackgroundPulmonary cysts and spontaneous pneumothorax are presented in most patients with Birt-Hogg-Dubé (BHD) syndrome, which is caused by loss of function mutations in the folliculin (FLCN) gene. The pathogenic mechanisms underlying the cystic lung disease in BHD are poorly understood.MethodsMesenchymal Flcn was specifically deleted in mice or in cultured lung mesenchymal progenitor cells using a Cre/loxP approach. Dynamic changes in lung structure, cellular and molecular phenotypes and signalling were measured by histology, immunofluorescence staining and immunoblotting.ResultsDeletion of Flcn in mesoderm-derived mesenchymal cells results in significant reduction of postnatal alveolar growth and subsequent alveolar destruction, leading to cystic lesions. Cell proliferation and alveolar myofibroblast differentiation are inhibited in the Flcn knockout lungs, and expression of the extracellular matrix proteins Col3a1 and elastin are downregulated. Signalling pathways including mTORC1, AMP-activated protein kinase, ERK1/2 and Wnt-β-catenin are differentially affected at different developmental stages. All the above changes have statistical significance (p<0.05).ConclusionsMesenchymal Flcn is an essential regulator during alveolar development and maintenance, through multiple cellular and molecular mechanisms. The mesenchymal Flcn knockout mouse model provides the first in vivo disease model that may recapitulate the stages of cyst development in human BHD. These findings elucidate the developmental origins and mechanisms of lung disease in BHD.


2018 ◽  
Vol 49 (4) ◽  
pp. 1659-1676 ◽  
Author(s):  
Xudong Peng ◽  
Qingjie Kang ◽  
Rui Wan ◽  
Ziwei Wang

Background/Aims: Previous studies demonstrated that HOXC9 acts as an oncogene in several tumors. The aim of this study was to explore whether HOXC9 promotes gastric cancer (GC) progression and elucidate the underlying molecular mechanisms. Methods: HOXC9 expression in GC tissues and adjacent non-cancer tissues was detected by quantitative RT-PCR (qRT-PCR) and immunohistochemistry. The functional effects of HOXC9 on proliferation, metastasis and stem cell-like phenotype were evaluated by relevant experiments in GC cells. The effect of miR-26a on HOXC9 was investigated by gain- and loss-of-function assays and luciferase reporter assay. Nude mouse models were established to test the effect of miR-26a and HOXC9 on tumorigenesis and metastasis of GC cells in vivo. Results: Herein, we showed that HOXC9 was upregulated in GC tissues and associated with a poor prognosis. HOXC9 knockdown inhibited the metastasis and stem cell-like phenotype of GC cells without significant effects on cell proliferation. In addition, we identifed HOXC9 as a direct target of miR-26a. Restoration of miR-26a in GC cells downregulated HOXC9 and reversed its promoting effect on metastasis and self-renewal, whereas miR-26a silencing upregulated HOXC9. In vivo experiments showed that HOXC9 knockdown suppressed tumorigenesis and lung metastasis of GC cells in nude mice, and these effects were mimicked by restoration of miR-26a. Conclusion: The present study demonstrates that HOXC9 promotes the metastasis and stem cell-like phenotype of GC cells, and this phenomenon can be reversed by restoration of miR-26a.


2021 ◽  
Author(s):  
Xinqiang Zhu ◽  
Xuetong Jiang ◽  
Qinglin Zhang ◽  
Hailong Huang ◽  
Xiaohong Shi ◽  
...  

Abstract Background: This study aimed to investigate the biological function and regulatory mechanism of TCN1 in colorectal cancer (CRC). Methods: We studied the biological functions of TCN1 using gain-of-function and loss-of-function analysis in HCT116 cell lines, and examined the effects of TCN1 on the proliferation, apoptosis, and invasion of CRC cells and determined its potential molecular mechanisms using CRC lines and mouse xenotransplantation models. Tumor xenograft and tumor metastasis studies were performed to detect the tumorigenicity and metastasis of cells in vivo. Results: TCN1-knockdown attenuated CRC cell proliferation, invasion and promoted cell apoptosis. Overexpression of TCN1 yielded the opposite effects. In addition, TCN1-knockdown HCT116 cells failed to form metastatic foci in the peritoneum after intravenous injection. Molecular mechanism studies showed that TCN1 interacts with integrin subunit β4 (ITGB4) to positively regulate the expression of ITGB4. TCN1-knockdown promoted the degradation of ITGB4 and increased the instability of ITGB4 and filamin A (FLNA). Downregulation of ITGB4 at the protein level resulted in the disassociation of the ITGB4/PLEC complex, leading to cytoskeletal damage. Conclusion: TCN1 might exert oncogenic role in CRC via regulating the ITGB4 signaling pathway.


Author(s):  
Marco Giordano ◽  
Alessandra Decio ◽  
Chiara Battistini ◽  
Micol Baronio ◽  
Fabrizio Bianchi ◽  
...  

Abstract Background Cancer stem cells (CSC) have been implicated in tumor progression. In ovarian carcinoma (OC), CSC drive tumor formation, dissemination and recurrence, as well as drug resistance, thus contributing to the high death-to-incidence ratio of this disease. However, the molecular basis of such a pathogenic role of ovarian CSC (OCSC) has been elucidated only to a limited extent. In this context, the functional contribution of the L1 cell adhesion molecule (L1CAM) to OC stemness remains elusive. Methods The expression of L1CAM was investigated in patient-derived OCSC. The genetic manipulation of L1CAM in OC cells provided gain and loss-of-function models that were then employed in cell biological assays as well as in vivo tumorigenesis experiments to assess the role of L1CAM in OC cell stemness and in OCSC-driven tumor initiation. We applied antibody-mediated neutralization to investigate L1CAM druggability. Biochemical approaches were then combined with functional in vitro assays to study the molecular mechanisms underlying the functional role of L1CAM in OCSC. Results We report that L1CAM is upregulated in patient-derived OCSC. Functional studies showed that L1CAM promotes several stemness-related properties in OC cells, including sphere formation, tumor initiation and chemoresistance. These activities were repressed by an L1CAM-neutralizing antibody, pointing to L1CAM as a druggable target. Mechanistically, L1CAM interacted with and activated fibroblast growth factor receptor-1 (FGFR1), which in turn induced the SRC-mediated activation of STAT3. The inhibition of STAT3 prevented L1CAM-dependent OC stemness and tumor initiation. Conclusions Our study implicate L1CAM in the tumorigenic function of OCSC and point to the L1CAM/FGFR1/SRC/STAT3 signaling pathway as a novel driver of OC stemness. We also provide evidence that targeting this pathway can contribute to OC eradication.


2020 ◽  
Vol 13 (10) ◽  
pp. dmm045815 ◽  
Author(s):  
Gideon L. Hughes ◽  
Michael A. Lones ◽  
Matthew Bedder ◽  
Peter D. Currie ◽  
Stephen L. Smith ◽  
...  

ABSTRACTAnimal models of human disease provide an in vivo system that can reveal molecular mechanisms by which mutations cause pathology, and, moreover, have the potential to provide a valuable tool for drug development. Here, we have developed a zebrafish model of Parkinson's disease (PD) together with a novel method to screen for movement disorders in adult fish, pioneering a more efficient drug-testing route. Mutation of the PARK7 gene (which encodes DJ-1) is known to cause monogenic autosomal recessive PD in humans, and, using CRISPR/Cas9 gene editing, we generated a Dj-1 loss-of-function zebrafish with molecular hallmarks of PD. To establish whether there is a human-relevant parkinsonian phenotype in our model, we adapted proven tools used to diagnose PD in clinics and developed a novel and unbiased computational method to classify movement disorders in adult zebrafish. Using high-resolution video capture and machine learning, we extracted novel features of movement from continuous data streams and used an evolutionary algorithm to classify parkinsonian fish. This method will be widely applicable for assessing zebrafish models of human motor diseases and provide a valuable asset for the therapeutics pipeline. In addition, interrogation of RNA-seq data indicate metabolic reprogramming of brains in the absence of Dj-1, adding to growing evidence that disruption of bioenergetics is a key feature of neurodegeneration.This article has an associated First Person interview with the first author of the paper.


2019 ◽  
Vol 21 (Supplement_6) ◽  
pp. vi263-vi263
Author(s):  
Rick Heinz ◽  
Sheri Holmen

Abstract Gliomas are the most common primary central nervous system malignancy in adults, but the molecular mechanisms responsible for their development and progression are not fully understood. Recent genomic analysis of World Health Organization grade II-III gliomas identifies 3 molecular subtypes of low grade glioma: no IDH mutation; IDH mutation without 1p/19q co-deletion; and IDH mutation with 1p/19q co-deletion. The latter, categorized as oligodendroglioma, commonly expresses loss of function mutations in CIC, FUBP1, and activation of PIK3CA. However, it is unknown if any of these mutations are sufficient to promote glioma development in cooperation with mutant IDH. Furthermore, research in oligodendroglioma is hampered by the lack of in vivo models of this specific glioma subtype. By utilizing the established RCAS/TVA somatic cell gene delivery method, mutant IDH1 can be expressed in the brains of mice. The tumorgenicity of other mutated genes associated with oligodendroglioma can be determined using additional methods such as conditional gene knockout and in vivo CRISPR-Cas9 mediated deletion. A mouse model for oligodendroglioma may identify new targetable genetic drivers of oligodendroglioma that will be useful for testing novel therapeutic strategies.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 1595-1595
Author(s):  
Weili Sun ◽  
Shirley Steward ◽  
Tamara Pestina ◽  
Carl W. Jackson ◽  
James R. Downing

Abstract The AML1/CBFβ transcription complex, a critical regulator of the formation of definitive hematopoietic stem cells (HSC), is one of the most frequent targets of genetic alterations in acute leukemia. In addition to somatic alterations of AML1 and CBFβ in acute leukemia, germ-line loss-of-function mutations of AML1 are the underlying cause of an autosomal dominant familial platelet disorder with a predisposition to acute myeloid leukemia (FPD/AML). Importantly, a subset of the mutations identified in families with FPD/AML result in AML1 null allele, suggesting that AML1 haploinsufficiency is the underlying molecular abnormality. To explore the functional consequences of AML1 halpoinsufficiency on megakaryocyte development and platelet function, we analyzed the hematopoietic system of AML1+/- mice. Loss of a single AML1 allele resulted in a 15% reduction in the number of circulating platelets and a significant impairment in platelet function including a decrease in dense granule content and an impaired ability to aggregate in response to collagen stimulation. Further analysis indentified a left shift in the DNA ploidy of megakaryocytes and a reduction in GPV expression, consistent with impaired megakaryocyte maturation. In addition, electron microscopy indicated a reduction in platelet demarcation channels within the cytoplasm of megakaryocytes. Importantly, however, we did not observe a reduction in the total number of megakaryoctyes or a decrease in megakaryocyte colony forming units. These data suggest that the haploinsufficiency of AML1+/− does not alter the initial formation of megakaryocytes, but instead impairs the ability of these cells to efficiently mature and produce functional platelets. To explore the underlying mechanism responsible for the observed impairment in megakaryocyte maturation, we analyzed the pattern of expression of several putative AML1 transcriptional targets. Although AML1 binding sites have been identified within the promoter of c-mpl, the gene encoding the receptor for thrombopoietin (TPO), we did not observe any difference in c-mpl expression levels or in circulating TPO concentration between AML1+/− and +/+ mice. In addition, in vivo TPO stimulation induced a similar magnitude of megakaryocyte maturation and platelet production in both AML1+/+ and +/− mice. By contrast, analysis of members of the protein kinase C (PKC) family of gene, several which have been identified as transcriptional targets of AML1, revealed a reduction in PKCδ levels in platelets from AML1+/− mice. Taken together, our data suggest that AML1 haploinsufficiency leads to abnormalities in platelet that are identical to those observed in patients with FPD/AML. Thus, these mice should prove useful for exploring the molecular mechanisms through which AML regulates the normal maturation of megakaryocytes. Our early analysis suggests altered PKCδ signaling is a possible contributing factor to the observed phenotypic abnormalities.


2013 ◽  
Vol 288 (20) ◽  
pp. 14531-14543 ◽  
Author(s):  
Tao Sun ◽  
Nuo Yu ◽  
Lu-Kai Zhai ◽  
Na Li ◽  
Chao Zhang ◽  
...  

The development of neuronal polarity is essential for the establishment of the accurate patterning of neuronal circuits in the brain. However, little is known about the underlying molecular mechanisms that control rapid axon elongation during neuronal development. Here, we report that c-Jun NH2-terminal kinase (JNK)-interacting protein-3 (JIP3) is highly expressed at axon tips during the critical period for axon development. Using gain- and loss-of-function approaches, immunofluorescence analysis, and in utero electroporation, we find that JIP3 can enhance axon elongation in primary hippocampal neurons and cortical neurons in vivo. We further demonstrate that JIP3 promotes axon elongation in a kinesin- and JNK-dependent manner using several deletion mutants of JIP3. Next, we demonstrate that the successful transportation of JIP3 to axon tips by kinesin is a prerequisite for enhancing JNK phosphorylation in this area and therefore promotes axon elongation, constituting a novel mechanism for coupling JIP3 anterograde transport with JNK signaling at the distal axons and axon elongation. Finally, our immunofluorescence data suggest that the activation of JNK at axon tips facilitates axon elongation by modulating cofilin activity and actin filament dynamics. These findings may have important implications for our understanding of neuronal axon elongation during development.


2012 ◽  
Vol 197 (4) ◽  
pp. 509-521 ◽  
Author(s):  
Jianwen Wei ◽  
Yu Shi ◽  
Lihua Zheng ◽  
Bin Zhou ◽  
Hiroyuki Inose ◽  
...  

A screen of microRNAs preferentially expressed in osteoblasts identified members of the miR-34 family as regulators of osteoblast proliferation and/or differentiation. Osteoblast-specific gain- and loss-of-function experiments performed in vivo revealed that miR-34b and -c affected skeletogenesis during embryonic development, as well as bone mass accrual after birth, through two complementary cellular and molecular mechanisms. First, they inhibited osteoblast proliferation by suppressing Cyclin D1, CDK4, and CDK6 accumulation. Second, they inhibited terminal differentiation of osteoblasts, at least in part through the inhibition of SATB2, a nuclear matrix protein that is a critical determinant of osteoblast differentiation. Genetic evidence obtained in the mouse confirmed the importance of SATB2 regulation by miR-34b/c. These results are the first to identify a family of microRNAs involved in bone formation in vivo and to identify a specific genetic pathway by which these microRNAs regulate osteoblast differentiation.


Sign in / Sign up

Export Citation Format

Share Document