Next Article in Journal
Molecular Characterization, Purification, and Mode of Action of Enterocin KAE01 from Lactic Acid Bacteria and Its In Silico Analysis against MDR/ESBL Pseudomonas aeruginosa
Previous Article in Journal
Identifying the Genetic Basis of Mineral Elements in Rice Grain Using Genome-Wide Association Mapping
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Diverse Role of CUB and Sushi Multiple Domains 1 (CSMD1) in Human Diseases

Division of Molecular Medicine, Leeds Institute of Medical Research, St James’s University Hospital, University of Leeds, Leeds LS9 7TF, UK
*
Author to whom correspondence should be addressed.
Genes 2022, 13(12), 2332; https://doi.org/10.3390/genes13122332
Submission received: 19 October 2022 / Revised: 1 December 2022 / Accepted: 2 December 2022 / Published: 10 December 2022
(This article belongs to the Section Molecular Genetics and Genomics)

Abstract

:
CUB and Sushi Multiple Domains 1 (CSMD1), a tumour suppressor gene, encodes a large membrane-bound protein including a single transmembrane domain. This transmembrane region has a potential tyrosine phosphorylation site, suggesting that CSMD1 is involved in controlling cellular functions. Although the specific mechanisms of action for CSMD1 have not yet been uncovered, it has been linked to a number of processes including development, complement control, neurodevelopment, and cancer progression. In this review, we summarise CSMD1 functions in the cellular processes involved in the complement system, metastasis, and Epithelial mesenchymal transition (EMT) and also in the diseases schizophrenia, Parkinson’s disease, and cancer. Clarifying the association between CSMD1 and the aforementioned diseases will contribute to the development of new diagnosis and treatment methods for these diseases. Recent studies in certain cancer types, e.g., gastric cancer, oesophageal cancer, and head and neck squamous cell carcinomas, have indicated the involvement of CSMD1 in response to immunotherapy.

Graphical Abstract

1. Introduction

Research on the CUB and Sushi Multiple Domains 1 (CSMD1) gene, which is thought to be associated with tumour suppression and the immune system, has increased recently. Initially, the CSMD1 gene was proven to be associated with schizophrenia (SZ), though CSMD1 is involved in many different mechanisms in the body with the discovery of its roles in the complement system, cancer, metastasis, cell migration, and epithelial–mesenchymal transition (EMT) [1,2,3]. Potentially, the CSMD1 gene may help clarify the unknown pathologies of many diseases in the future. Consequently, a good understanding of the structure and possible metabolism/mechanism of the CSMD1 protein is important. In this review, we provide an overview of the general properties of the CSMD1 gene and protein and its roles in various diseases.

2. CSMD1 Structure

The CUB and Sushi Multiple Domains 1 (CSMD1) gene consists of 71 exons and spans a 2 MB DNA region on chromosome 8p23.2 [4]. CSMD1 consists of 14 CUB domains separated by 14 complement control protein (CCP) domains at the N-terminus, followed by a tandem repetition of 15 CCP domains (Figure 1, [5]). The CSMD1 gene encodes 17 splice variants in total [1]. The largest of these variations codes for a 3508-amino-acid protein with a 383 kDa molecular mass [4]. Conservative CUB domains, which span 110 amino acids, were first discovered in a variety of extracellular proteins, the majority of which were known to play a role in developmental processes and immunity, as well as in various cancer types [6,7]. Sushi domains are extracellular motifs that are frequently found in protein–protein interactions. Every Sushi domain has ∼60 amino acid residues with conserved tryptophan, glycine, proline, hydrophobic residues, and four invariant cysteines. Since many complement control proteins contain multiple Sushi domains, it is also referred to as a complement control protein [8,9,10]. The CSMD1 protein is a membrane-bound protein including a single transmembrane domain [4]. A putative tyrosine phosphorylation site inside this transmembrane region suggests that CSMD1 is involved in the regulation of cellular processes [11,12,13]. According to the latest data based on a combination of internally generated Human Protein Atlas RNA-Seq data and RNA-Seq data from the Genotype-Tissue Expression (GTEx) project, CSDM1 is predominantly expressed in the epithelial cells in many tissues with high expression in the brain, male reproduction system, and retina, followed by the breast and female reproductive system, spleen, and kidney [14,15].

3. CSMD Gene Family

CSMD1 is currently the best understood protein in the CSMD protein family, consisting of three structurally very similar proteins, with 14 CUB domains separated by a sushi domain, an additional uninterrupted array of sushi domains, a single transmembrane domain, and a short cytoplasmic tail. CSMD2 and CSMD3 are expressed at low levels in many tissues with the highest expression in the central nervous system. This expression pattern is similar to that of CSMD1, though CSMD1 expression is lower than that of the other two genes [16]. The striking similarity of all three CSMD genes brought up the question of whether CSMD2 and 3 are also tumour suppressors. A colorectal cancer study revealed that all three CSMDs were lower in colorectal carcinoma cells than in normal tissues [17].
CSMD2 is located at the 1p34 chromosomal region, which may contain an oligodendroglioma suppressor, but its expression was found to be increased in some head and neck cancer cell lines [16]. Reduced CSMD2 expression was linked to tumour size, lymphatic invasion, and differentiation in colon cancer, the development of pulmonary sarcomatoid carcinoma, liver metastasis, and pancreatic cancer [17,18,19,20]. Furthermore, CSMD2 has a role in immature neuron development [21] and has also been identified as a risk factor for the development of schizophrenia (SZ) [22].
CSMD3, which is located on chromosome 8q23.3-q24.1, spans 1.2 Mb and encodes 73 exons [23]. CSMD3 expressed predominantly in the cortical neurons of the developing cortex was associated with neurodevelopmental disorders (NDDs) such as autism spectrum disorder (ASD) and SZ [24,25,26,27]. After elucidating the expression of CSMD3 in dendrites and its functions for dendrite formation [28], Song et al. proved that CSMD3 plays a critical role in early cortical neural network construction, which includes synaptogenesis and glia–vascular communication [29]. In addition, it was discovered that the CSMD3 mutation is strongly associated with increased tumour mutation burden and poor clinical prognosis in ovarian cancer [30]. Furthermore, common CSMD3 gene mutations were found in pulmonary carcinosarcomas [31] and oesophageal squamous cell carcinoma (OSCC) [32].

4. CSMD1 Function

CSMD1 has been implicated in a variety of processes including development, complement control, neurodevelopment disorders, and cancer progression, though specific mechanisms of action have yet to be identified (Figure 2, [5]) [3,33,34,35,36,37,38].
CSMD1 has been shown to modulate the SMAD pathway and has potential for intracellular signalling; due to its structural features, it may operate as a co-receptor or interact with growth factor receptors. It has also been reported that CSMD1 interacts with ECM components and goes through clathrin-mediated endocytosis (Figure 3, [5]) [39]. This review will focus on CSMD1 and its roles in the complement system, neurodevelopmental diseases, cancer and metastasis, and EMT.

4.1. CSMD1 in Complement System

The tandem Sushi domains at the C-terminus of CSMD1, a transmembrane protein composed of multiple CUB and Sushi repeats, have homology to the complementary activation of regulators (RCA) gene family [4,16,23,40]. Complement, a key part of the innate immune system, includes more than 30 proteins grouped in a proteolytic cascade, with effector mechanisms controlled by a variety of receptors [41]. Although complement is most plentiful in blood, all of its components can also be found in tissues due to diffusion or local transport [42]. Complement’s primary role is to recognise pathogens and damaged cells from an organism, induce inflammation, and destroy the pathogen’s cell membrane [43]. Complement is structured into three major pathways (classical, lectin, and alternative), each of which is inducted by sensory molecules that can detect pathogens or undesired material. This triggers a series of proteolytic activation processes that result in complement effector molecules such C3 fragments. The synthesis of the membrane attack complex (MAC), which can lyse Gram-negative bacteria and harm eukaryotic cells, is the last stage in the complement cascade [3]. The classical pathway is initiated by the binding of antibodies to antigens on the target cell. Complement C1s is activated as a result of this interaction, which is mediated via C1q activation. The C1s enzyme can cleave C4 and C2 components, resulting in the formation of C3 convertase (C4bC2b), which activates the complement cascade [41,44,45]. The alternative pathway stimulates the membrane attack complex by hydrolysing C3 molecules to produce a different C3 convertase (C3bBb). In this step, CSMD1 prevents C3 from being deposited on the cell surface, effectively shutting down the traditional complement pathway [37,40,46]. Consequently, CSMD1 was hypothesized to have a role in cell cycle regulation and apoptosis control due to its role on complement (Figure 4, [5]) [13].

4.2. CSMD1 in Neurodevelopment Diseases

The precise role of CSMD1 in immune responses awaits further elucidation, while a possible association of CSMD1 in autoimmune disease (neonatal lupus) has been reported [47]. In the light of current knowledge, we know that complement activity is tightly controlled in the brain and regulates C3/CR3-dependent axonal pruning by phagocytic microglia. During development, this mechanism maintains the perfect connection of neural circuits in the visual system [48]. Therefore, it is thought that complement may lead to abnormal synaptic elimination in other parts of the brain, which may affect the risk of both neurodegenerative and psychiatric disorders [49]. Furthermore, recent GWAS and SNP analysis have shown that CSMD1 is closely related to neurological diseases such as SZ and Parkinson’s disease (PD) [50,51].
The discovery of a Csmd1 promoter-associated lncRNA, which may be responsible for brain-specific promoter activity in the adult and developing central nervous system, added to the importance of Csmd1 for brain functioning [52]. Athanasiu et al. (2017) demonstrated that CSMD1 variants are associated with immediate episodic memory and cognitive function [22]. Along with a GWAS study, it suggested that CSMD1 is expressed significantly in the amygdala, which affects social behaviour, and CSMD1 can be used as a diagnostic marker for amygdala-related disorders [53].
In a study conducted with SZ patients, higher C4A mRNA levels were associated with more serious general psychopathology symptoms, while lower CSMD1 mRNA levels were predicted to cause worse working memory [54]. Some CSMD1 SNPs have been shown to be associated with cognitive ability in SZ patients [55]. It was also suggested that CSMD1 increased the risk of schizophrenia as a result of the negative effects of the A allele in CSMD1 rs10503253 on brain activity [56]. A 2021 study by Abd El Gayed et al. found that the CSMD1 mRNA expression and protein levels were significantly lower in SZ patients compared to the controls, suggesting that mRNA expression of the CSMD1 gene may be a reliable and early diagnostic predictor of first-episode SZ in familial high-risk Egyptian children and young adults [57].
According to the results of whole-exome sequencing analysis performed on two Spanish families diagnosed with PD, it has been suggested that mutations in the CSDM1 gene may cause the familial PD phenotype [58]. However, a study conducted on the Iranian population showed no association between CSMD1 rs12681349 polymorphism and PD [59]. A case-control study conducted on the Han population in northern China in 2021 showed that polymorphisms in the CSMD1 gene were closely related to PD, and significant differences were found in rs10503253 and rs1983474 polymorphisms between PD cases and controls. However, the researchers emphasized that studies in larger populations and other ethnic groups are needed to confirm the correlation between the CSMD1 polymorphism and PD [51].

4.3. CSMD1 in Cancer

Abnormalities of the short arm of chromosome 8 have been associated with many carcinomas’ pathogenesis [60]. Toomes et al. and other scientists showed a hemizygous or homozygous deletion of the chromosome band at 8p23 encoding the CSMD1 protein in different cancer types such as oropharyngeal squamous cell carcinomas (OSCCs) and head and neck squamous cell carcinomas (HNSCCs) [1,61,62,63]. Although the function of CSMD1 is unclear, previous studies have shown that this transmembrane protein is engaged in a signalling cascade that regulates a variety of cell processes implicated in cancer formation such as proliferation and cell migration [4,13,64]. In this review, the differential analysis of CSMD1 expression between normal and cancer tissues was determined based on RNA sequencing data using tnmplot [65]. This analysis showed that CSMD1 expression was significantly increased in acute myeloid leukaemia (AML), liver, pancreas, skin, thyroid, and uterine corpus (EC) endometrial carcinoma compared to controls. However, adrenal, bladder, breast, colon, ovary, prostate, rectum, renal chromophobe (CH) cell carcinoma, renal clear cell carcinoma (CC), renal papillary cell carcinoma (PA), and lung adenocarcinoma (AC) had less CSMD1 expression compared to controls (Figure 5, [65]).
Previously, we identified reduced CSMD1 expression in 79/275 (28.7%) of invasive ductal breast cancer patients, which were associated with high tumour grade and poor overall survival. More importantly, CSMD1 was also an independent predictor of overall survival [66]. An mRNA study by Escudero-Esparza et al. in 127 breast cancer samples also found that low levels of CSMD1 expression was associated with statistically significant lower survival compared to those with high levels [67]. In a later study, we created a three-dimensional culture model of MCF10A cells with decreased CSMD1 expression that indicated that the reduction of CSMD1 expression resulted in the formation of bigger and less differentiated breast ductal structures [35]. A recent deep whole-genome sequencing study identified CSMD1 deletions in 50% of breast cancer patient-derived xenografts, suggesting a role in driving aggressive breast cancer. The low expression of CSMD1 was also associated with reduced survival in the breast cancer METABRIC study [68]. Gialeli et al. suggested that CSMD1 provides tumour suppression by interacting with the EGFR pathway and can be used as a biomarker for predicting chemotherapy response in highly invasive breast cancer [69]. Additionally, Tang et al. discovered that the decreased expression of CSMD1 in melanoma cells has a lower influence on melanoma cell migration and proliferation, and that CSMD1 can serve as a tumour suppressor gene in melanoma cells [13]. Furthermore, Zhang et al. discovered that CSMD1 expression in colorectal cancer is low, is linked to overall survival, can be utilised as a predictor of colorectal cancer, and plays a vital part in the prognosis of the disease [17]. Additionally, reduced CSMD1 gene expression was associated with poor prognosis in HNSCC and prostate cancer [38,70]. A further HNSCC study interrogating three publicly available genome-wide expression datasets found CSMD1-inactived cancers demonstrated a reduced prognosis [12].
Similarly, Fan et al. showed that the CSMD1 mutation status was an independent predictor of prognosis in oesophageal cancer patients. This study also found CSMD1 wild-type oesophageal cancer patients were more susceptible to paclitaxel chemotherapy [71]. It has shown that CSMD1 is dramatically downregulated in gastric cancer (GC) tissues compared to normal tissues via RT-PCR, and the overexpression of microRNA-10b, a direct target of CSMD1 in GC cells, was found to increase GC cell vitality, migration, and invasion [72,73,74]. Another study discovered that miR-642b-3p acts as an oncomiR that promotes tumour progression in GC by repressing CSMD1 expression and inactivating the Smad signalling pathway, which could contribute to the development of potential therapies for GC treatment [75].
A study on hepatocellular carcinoma (HCC) identified lncCSMD1-1 upregulated in HCC and directly binding to the MYC protein in the nucleus of HCC cells, promoting HCC progression and the upregulation of MYC protein. Furthermore, lncCSMD1 has been shown to act as an oncogene, promoting HCC cell proliferation, migration, invasion, and EMT. As a result, it was speculated that lncCSMD1 could be a novel and reproducible prognostic biomarker for HCC patients, as well as playing an important role in HCC progression [76]. Using the GEO and TCGA databases, a study identified several novel driver genes including CSMD1 associated with HCC and demonstrated that this gene was strongly related to the prognosis of early recurrence and an effective prognostic marker for HCC [77].

4.4. CSMD1 in Metastasis and EMT

Metastasis is a complex process that includes events such as loss of adhesion, increased invasiveness, and motility to perform intravasation, joining the circulation through lymph nodes and blood vessels, and connecting to blood vessels [78]. Tumour cells transform from an epithelial phenotype to a mesenchymal phenotype to acquire invasive characteristics, and metastasis is one of the key points in the formation of this epithelial–mesenchymal transition (EMT). Significant evidence indicates that the tumour suppressor gene CSMD1 plays an important role in metastasis. Deletions of the CSDM1 locus chromosome 8p23 appear to be clinically important because an allele deficit at this coding region has been linked to poor prognosis, recurrence, and metastasis in many cancer types [79]. Previously, we demonstrated that CSMD1 knockdown using short hairpin RNA in three different cell lines (breast MCF10A, prostate LNCaP, and metastatic MDA-MB-435) resulted in striking morphological changes similar to those previously reported in EMT. Furthermore, functional assays on the MCF10A breast cancer line found loss of CSMD1 expression enhanced migration and invasion while reducing adhesion to Matrigel and fibronectin [35].
In melanoma cells, an increased expression of CSMD1 reduces the rate of cell migration [13]. In a xenograft model of human breast cancer, mice injected with MDA-MB-231 breast cancer cells, either alone or with artificially increased CSMD1 expression, found that there was a significant reduction in lung metastasis in the CSMD1-expressing group when compared to the controls. This study also demonstrated that the overexpression of CSMD1 in breast cancer cell lines BT20 and MCF7 inhibited migration, adhesion, and invasion [67].
EMT is a normal biological process driving mammary gland development. In order to investigate the association between CSMD1 and EMT further, mammary gland development during puberty was studied in a Csmd1 knockout (KO) mouse model. Our study identified increased ductal development during the early stages of puberty in the KO mice, characterised by increased ductal area and terminal end bud number at 6 weeks. The increased expression of various proteins (Stat1, Fak, Akt, Slug/Snail, and Progesterone receptor) was identified in the Csmd1 KO mice mammary glands at 4 weeks, followed by lower expression levels from 6 weeks in the KO mice compared to the wild-type control mice. This study found an association between Csmd1 and cell invasiveness regulation, which might be regulated by changes in cell adhesion processes (Figure 6, [5]) [33]. Our study indicates a novel role for Csmd1 in mammary gland development, with Csmd1 KO causing significantly more rapid mammary gland development, suggesting an earlier adult mammary gland formation [33].

4.5. CSMD1 Inactivation

There are many different ways that CSMD1 might be dysregulated, including epigenetic silencing, CSMD1 gene deletions and mutations, and microRNA interference. Interrogation of the Cancer Genome Atlas (TCGA) has identified alterations in CSMD1 in many malignant tumours such as breast cancer (~5%), prostate cancer (5.7%), bladder urothelial carcinoma (8%), lung cancer (7%), ovarian cancer (7%), liver cancer (7%), and colorectal adenocarcinoma (7.4%) (Figure 2). It was discovered that miR-10b reduces the expression of CSMD1 and that it is substantially expressed in HCC compared to healthy tissue [64]. Similarly, the downregulation of CSMD1 in glioblastoma was associated with the overexpression of miRNA-10a and miRNA-10b [80]. MiR-137, which was discovered through genome-wide association studies in SZ, is another microRNA that specifically targets CSMD1 [81]. Both miR-10b and miR-137 have been found to bind to the 3′ untranslated region of the CSMD1 gene [80,81].

4.6. CSMD1 and Immunotherapy

Recently, several studies have suggested that CSMD1-inactivated cancers may respond to immune checkpoint inhibitors [12,71,72,73]. An oesophageal cancer study found CSMD1 mutated cancers were associated with a high tumour mutation burden (TMB). As expected, the high TMB correlated with high expression of PD-1, suggesting that these patients may benefit from PD-1 inhibitor immunotherapy [71]. Furthermore, a large-scale genomic study on HNSCC cancers demonstrated an association between CSMD1-inactivation and tumour immunity [12]. Similarly, a GC study revealed that patients with CSMD1 mutations had significantly higher TMB and better prognoses than CSMD1-wild patients. Following that, it showed that several immune-related signalling pathways were upregulated in the CSMD1-mut samples; that there was a higher proportion of anti-tumour immune cells, such as CD4+ Th1 cells, NK cells, M1 macrophage cells, and PDC; a lower proportion of tumour-promoting immune cells, such as Treg cells, M2 macrophage cells, and endothelial cells; and that PD-L1 was upregulated [73]. Another GC study confirmed the association between CSMD1 mutation with TMB and high PDL1 expression and increased survival supporting the potential of CSMD1 as a biomarker for assessing immune checkpoint inhibitor therapy in GC patients [74].

5. Conclusions

Elucidating the relationship of CSMD1 with the aforementioned diseases in this review may be important in terms of both treatment and diagnosis in the future. Recent studies have revealed a possible role for CSMD1 in immunotherapy for some cancer types [73,74,75,76,77,78,79,80,81,82]. The determination of the factors affecting the expression of CSMD1 and the clarification of the signalling pathways it affects will increase the clinical utility of CSMD1.

Author Contributions

Original draft preparation, E.E.A. and S.M.B.; review and editing, E.E.A. and S.M.B. All authors have read and agreed to the published version of the manuscript.

Funding

E.E.A. was supported by a scholarship from the Turkish Ministry of National Education, Republic of Turkey. S.M.B. was supported by the University of Leeds.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

All relevant data have been cited within this review article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Toomes, C.; Jackson, A.; Maguire, K.; Wood, J.; Gollin, S.; Ishwad, C.; Paterson, I.; Prime, S.; Parkinson, K.; Bell, S. The presence of multiple regions of homozygous deletion at the CSMD1 locus in oral squamous cell carcinoma question the role of CSMD1 in head and neck carcinogenesis. Genes Chromosomes Cancer 2003, 37, 132–140. [Google Scholar] [CrossRef] [PubMed]
  2. Baum, M.L.; Wilton, D.K.; Muthukumar, A.; Fox, R.G.; Carey, A.; Crotty, W.; Scott-Hewitt, N.; Bien, E.; Sabatini, D.A.; Lanser, T. CUB and Sushi Multiple Domains 1 (CSMD1) opposes the complement cascade in neural tissues. bioRxiv 2020. [Google Scholar] [CrossRef]
  3. Blom, A. The role of complement inhibitors beyond controlling inflammation. J. Intern. Med. 2017, 282, 116–128. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Sun, P.C.; Uppaluri, R.; Schmidt, A.P.; Pashia, M.E.; Quant, E.C.; Sunwoo, J.B.; Gollin, S.M.; Scholnick, S.B. Transcript map of the 8p23 putative tumor suppressor region. Genomics 2001, 75, 17–25. [Google Scholar] [CrossRef]
  5. Created with BioRender.com. 2022. Available online: https://app.biorender.com (accessed on 6 June 2022).
  6. Bork, P.; Beckmann, G. The CUB domain: A widespread module in developmentally regulated proteins. J. Mol. Biol. 1993, 231, 539–545. [Google Scholar] [CrossRef]
  7. Gaboriaud, C.; Gregory-Pauron, L.; Teillet, F.; Thielens, N.M.; Bally, I.; Arlaud, G.J. Structure and properties of the Ca2+-binding CUB domain, a widespread ligand-recognition unit involved in major biological functions. Biochem. J. 2011, 439, 185–193. [Google Scholar] [CrossRef] [Green Version]
  8. Ichinose, A.; Bottenus, R.E.; Davie, E.W. Structure of transglutaminase. J. Biol. Chem. 1990, 265, 13411–13414. [Google Scholar] [CrossRef]
  9. Kirkitadze, M.D.; Barlow, P.N. Structure and flexibility of the multiple domain proteins that regulate complement activation. Immunol. Rev. 2001, 180, 146–161. [Google Scholar] [CrossRef]
  10. Reid, K.B.; Day, A.J. Structure-function relationships of the complement components. Immunol. Today 1989, 10, 177–180. [Google Scholar] [CrossRef]
  11. Blom, N.; Gammeltoft, S.; Brunak, S. Sequence and structure-based prediction of eukaryotic protein phosphorylation sites. J. Mol. Biol. 1999, 294, 1351–1362. [Google Scholar] [CrossRef]
  12. Jung, A.R.; Eun, Y.-G.; Lee, Y.C.; Noh, J.K.; Kwon, K.H. Clinical significance of CUB and sushi multiple domains 1 inactivation in head and neck squamous cell carcinoma. Int. J. Mol. Sci. 2018, 19, 3996. [Google Scholar] [CrossRef] [Green Version]
  13. Tang, M.-R.; Wang, Y.-X.; Guo, S.; Han, S.-Y.; Wang, D. CSMD1 exhibits antitumor activity in A375 melanoma cells through activation of the Smad pathway. Apoptosis 2012, 17, 927–937. [Google Scholar] [CrossRef] [PubMed]
  14. The Human Protein Atlas. CSMD1. Available online: https://www.proteinatlas.org/ENSG00000183117-CSMD1/tissue (accessed on 6 June 2022).
  15. Uhlén, M.; Fagerberg, L.; Hallström, B.M.; Lindskog, C.; Oksvold, P.; Mardinoglu, A.; Sivertsson, Å.; Kampf, C.; Sjöstedt, E.; Asplund, A.; et al. Tissue-based map of the human proteome. Science 2015, 347, 1260419. [Google Scholar] [CrossRef] [PubMed]
  16. Lau, W.L.; Scholnick, S.B. Identification of two new members of the CSMD gene family☆. Genomics 2003, 82, 412–415. [Google Scholar] [CrossRef] [PubMed]
  17. Zhang, R.; Song, C. Loss of CSMD1 or 2 may contribute to the poor prognosis of colorectal cancer patients. Tumor Biol. 2014, 35, 4419–4423. [Google Scholar] [CrossRef]
  18. Yang, P.-S.; Hsu, H.-H.; Hsu, T.-C.; Chen, M.-J.; Wang, C.-D.; Yu, S.-L.; Hsu, Y.-C.; Li, K.-C. Genome-wide scan for copy number alteration association with relapse-free survival in colorectal cancer with liver metastasis patients. J. Clin. Med. 2018, 7, 446. [Google Scholar] [CrossRef] [Green Version]
  19. Yan, J.; Li, S.; Xi, W.; Chen, D.; Xiao, M. Immunoregulation related gene of CSMD2 and RNA activity as potential predicter of 2-year survival in pancreatic cancer: A DNA and RNA sequencing analysis. J. Clin. Oncol. 2021, 39, e16212. [Google Scholar] [CrossRef]
  20. Zhang, C.; Li, Z.; Zhang, Y.; Zhao, C.; Wang, H.; Lin, J.; Liu, C.; Wang, X.; Wang, H. Genomic Variations and Immune-Related Features of TMB, PD-L1 Expression and CD8+ T Cell Infiltration in Chinese Pulmonary Sarcomatoid Carcinoma. Int. J. Gen. Med. 2022, 15, 4209. [Google Scholar] [CrossRef]
  21. Gutierrez, M.A.; Dwyer, B.E.; Franco, S.J. Csmd2 Is a Synaptic Transmembrane Protein that Interacts with PSD-95 and Is Required for Neuronal Maturation. Eneuro 2019, 6. [Google Scholar] [CrossRef] [Green Version]
  22. Athanasiu, L.; Giddaluru, S.; Fernandes, C.; Christoforou, A.; Reinvang, I.; Lundervold, A.J.; Nilsson, L.-G.; Kauppi, K.; Adolfsson, R.; Eriksson, E. A genetic association study of CSMD1 and CSMD2 with cognitive function. Brain Behav. Immun. 2017, 61, 209–216. [Google Scholar] [CrossRef]
  23. Shimizu, A.; Asakawa, S.; Sasaki, T.; Yamazaki, S.; Yamagata, H.; Kudoh, J.; Minoshima, S.; Kondo, I.; Shimizu, N. A novel giant gene CSMD3 encoding a protein with CUB and sushi multiple domains: A candidate gene for benign adult familial myoclonic epilepsy on human chromosome 8q23. 3–q24. 1. Biochem. Biophys. Res. Commun. 2003, 309, 143–154. [Google Scholar] [CrossRef] [PubMed]
  24. Wu, J.; Yu, P.; Jin, X.; Xu, X.; Li, J.; Li, Z.; Wang, M.; Wang, T.; Wu, X.; Jiang, Y. Genomic landscapes of Chinese sporadic autism spectrum disorders revealed by whole-genome sequencing. J. Genet. Genom. 2018, 45, 527–538. [Google Scholar] [CrossRef] [PubMed]
  25. Curran, S.; Ahn, J.W.; Grayton, H.; Collier, D.A.; Ogilvie, C.M. NRXN1 deletions identified by array comparative genome hybridisation in a clinical case series–further understanding of the relevance of NRXN1 to neurodevelopmental disorders. J. Mol. Psychiatry 2013, 1, 4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Magri, C.; Sacchetti, E.; Traversa, M.; Valsecchi, P.; Gardella, R.; Bonvicini, C.; Minelli, A.; Gennarelli, M.; Barlati, S. New copy number variations in schizophrenia. PLoS ONE 2010, 5, e13422. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Malhotra, D.; McCarthy, S.; Michaelson, J.J.; Vacic, V.; Burdick, K.E.; Yoon, S.; Cichon, S.; Corvin, A.; Gary, S.; Gershon, E.S. High frequencies of de novo CNVs in bipolar disorder and schizophrenia. Neuron 2011, 72, 951–963. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Mizukami, T.; Kohno, T.; Hattori, M. CUB and Sushi multiple domains 3 regulates dendrite development. Neurosci. Res. 2016, 110, 11–17. [Google Scholar] [CrossRef]
  29. Song, W.; Li, Q.; Wang, T.; Li, Y.; Fan, T.; Zhang, J.; Wang, Q.; Pan, J.; Dong, Q.; Sun, Z.S. Putative complement control protein CSMD3 dysfunction impairs synaptogenesis and induces neurodevelopmental disorders. Brain Behav. Immun. 2022, 102, 237–250. [Google Scholar] [CrossRef] [PubMed]
  30. Lu, N.; Liu, J.; Xu, M.; Liang, J.; Wang, Y.; Wu, Z.; Xing, Y.; Diao, F. CSMD3 is Associated with Tumor Mutation Burden and Immune Infiltration in Ovarian Cancer Patients. Int. J. Gen. Med. 2021, 14, 7647. [Google Scholar] [CrossRef]
  31. Li, F.; Hu, S.; Kong, K.; Cao, P.; Han, P.; Deng, Y.; Zhao, B. Next-generation sequencing analysis identified genomic alterations in pathological morphologies of 3 cases of pulmonary carcinosarcoma. OncoTargets Ther. 2020, 13, 7963. [Google Scholar] [CrossRef]
  32. Yang, H.-S.; Liu, W.; Zheng, S.-Y.; Cai, H.-Y.; Luo, H.-H.; Feng, Y.-F.; Lei, Y.-Y. A Novel Ras--Related Signature Improves Prognostic Capacity in Oesophageal Squamous Cell Carcinoma. Front. Genet. 2022, 13, 822966. [Google Scholar] [CrossRef]
  33. Burgess, S.J.; Gibbs, H.; Toomes, C.; Coletta, P.L.; Bell, S.M. The role of Csmd1 during mammary gland development. Genes 2021, 12, 162. [Google Scholar] [CrossRef] [PubMed]
  34. Lee, A.S.; Rusch, J.; Lima, A.C.; Usmani, A.; Huang, N.; Lepamets, M.; Vigh-Conrad, K.A.; Worthington, R.E.; Mägi, R.; Wu, X. Rare mutations in the complement regulatory gene CSMD1 are associated with male and female infertility. Nat. Commun. 2019, 10, 4626. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Kamal, M.; Holliday, D.L.; Morrison, E.E.; Speirs, V.; Toomes, C.; Bell, S.M. Loss of CSMD1 expression disrupts mammary duct formation while enhancing proliferation, migration and invasion. Oncol. Rep. 2017, 38, 283–292. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Donohoe, G.; Walters, J.; Hargreaves, A.; Rose, E.J.; Morris, D.; Fahey, C.; Bellini, S.; Cummins, E.; Giegling, I.; Hartmann, A.M. Neuropsychological effects of the CSMD1 genome-wide associated schizophrenia risk variant rs10503253. Genes Brain Behav. 2013, 12, 203–209. [Google Scholar] [CrossRef]
  37. Escudero-Esparza, A.; Kalchishkova, N.; Kurbasic, E.; Jiang, W.G.; Blom, A.M. The novel complement inhibitor human CUB and Sushi multiple domains 1 (CSMD1) protein promotes factor I-mediated degradation of C4b and C3b and inhibits the membrane attack complex assembly. FASEB J. 2013, 27, 5083–5093. [Google Scholar] [CrossRef]
  38. Ma, C.; Quesnelle, K.M.; Sparano, A.; Rao, S.; Park, M.S.; Cohen, M.A.; Wang, Y.; Samanta, M.; Kumar, M.S.; Aziz, M.U. Characterization CSMD1 in a large set of primary lung, head and neck, breast and skin cancer tissues. Cancer Biol. Ther. 2009, 8, 907–916. [Google Scholar] [CrossRef] [Green Version]
  39. Gialeli, C.; Gungor, B.; Blom, A.M. Novel potential inhibitors of complement system and their roles in complement regulation and beyond. Mol. Immunol. 2018, 102, 73–83. [Google Scholar] [CrossRef]
  40. Kraus, D.M.; Elliott, G.S.; Chute, H.; Horan, T.; Pfenninger, K.H.; Sanford, S.D.; Foster, S.; Scully, S.; Welcher, A.A.; Holers, V.M. CSMD1 is a novel multiple domain complement-regulatory protein highly expressed in the central nervous system and epithelial tissues. J. Immunol. 2006, 176, 4419–4430. [Google Scholar] [CrossRef] [Green Version]
  41. Ricklin, D.; Hajishengallis, G.; Yang, K.; Lambris, J.D. Complement: A key system for immune surveillance and homeostasis. Nat. Immunol. 2010, 11, 785–797. [Google Scholar] [CrossRef] [Green Version]
  42. Martin, M.; Blom, A.M. Complement in removal of the dead–balancing inflammation. Immunol. Rev. 2016, 274, 218–232. [Google Scholar] [CrossRef]
  43. Sarma, J.V.; Ward, P.A. The complement system. Cell Tissue Res. 2011, 343, 227–235. [Google Scholar] [PubMed]
  44. Merle, N.S.; Church, S.E.; Fremeaux-Bacchi, V.; Roumenina, L.T. Complement system part I–molecular mechanisms of activation and regulation. Front. Immunol. 2015, 6, 262. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Garcia, B.L.; Zwarthoff, S.A.; Rooijakkers, S.H.; Geisbrecht, B.V. Novel evasion mechanisms of the classical complement pathway. J. Immunol. 2016, 197, 2051–2060. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Håvik, B.; Le Hellard, S.; Rietschel, M.; Lybæk, H.; Djurovic, S.; Mattheisen, M.; Mühleisen, T.W.; Degenhardt, F.; Priebe, L.; Maier, W. The complement control-related genes CSMD1 and CSMD2 associate to schizophrenia. Biol. Psychiatry 2011, 70, 35–42. [Google Scholar] [CrossRef] [PubMed]
  47. Ramos, P.S.; Marion, M.C.; Langefeld, C.D.; Genetics, I.C.o.S.L.E.; Buyon, J.P.; Clancy, R.M. Brief report: Enrichment of associations in genes with fibrosis, apoptosis, and innate immunity functions with cardiac manifestations of neonatal lupus. Arthritis Rheum. 2012, 64, 4060–4065. [Google Scholar] [CrossRef] [Green Version]
  48. Schafer, D.; Lehrman, E.; Kautzman, A.; Koyama, R.; Mardinly, A.; Yamasaki, R.; Ransohoff, R.; Greenberg, M.; Barres, B.; Stevens, B. Microglia sculpt postnatal neural circuits in an activity and complement-dependent manner. Neuron 2012, 74, 691–705. [Google Scholar] [CrossRef] [Green Version]
  49. Schafer, D.P.; Lehrman, E.K.; Stevens, B. The “quad-partite” synapse: Microglia-synapse interactions in the developing and mature CNS. Glia 2013, 61, 24–36. [Google Scholar] [CrossRef] [Green Version]
  50. Ripke, S.; Sanders, A.; Kendler, K.; Levinson, D.; Sklar, P.; Holmans, P.; Lin, D.; Duan, J.; Ophoff, R.; Andreassen, O. Schizophrenia Psychiatric Genome-Wide Association Study (GWAS) Consortium Genome-wide association study identifies five new schizophrenia loci. Nat. Genet. 2011, 43, 969–976. [Google Scholar]
  51. Bai, X.; Jin, J.; Li, S.; Wang, H.; Xie, A. CUB and sushi Multiple domains (CSMD1) gene polymorphisms and susceptibilities to idiopathic Parkinson’s disease in northern Chinese Han population: A case-control study. Parkinson’s Dis. 2021, 2021, 6661162. [Google Scholar] [CrossRef]
  52. Steen, V.M.; Nepal, C.; Ersland, K.M.; Holdhus, R.; Nævdal, M.; Ratvik, S.M.; Skrede, S.; Håvik, B. Neuropsychological deficits in mice depleted of the schizophrenia susceptibility gene CSMD1. PLoS ONE 2013, 8, e79501. [Google Scholar] [CrossRef]
  53. Bickart, K.; Napolioni, V.; Khan, R.; Kim, Y.; Altmann, A.; Richiardi, J.; Newsom, M.; Sadaghiani, S.; Banaschewski, T.; Bokde, A. Genetic variation in CSMD1 affects amygdala connectivity and prosocial behavior. bioRxiv 2020. [Google Scholar] [CrossRef]
  54. Hatzimanolis, A.; Foteli, S.; Stefanatou, P.; Ntigrintaki, A.-A.; Ralli, I.; Kollias, K.; Nikolaou, C.; Gazouli, M.; Stefanis, N.C. Deregulation of complement components C4A and CSMD1 peripheral expression in first-episode psychosis and links to cognitive ability. Eur. Arch. Psychiatry Clin. Neurosci. 2022, 272, 1219–1228. [Google Scholar] [CrossRef] [PubMed]
  55. Koiliari, E.; Roussos, P.; Pasparakis, E.; Lencz, T.; Malhotra, A.; Siever, L.J.; Giakoumaki, S.G.; Bitsios, P. The CSMD1 genome-wide associated schizophrenia risk variant rs10503253 affects general cognitive ability and executive function in healthy males. Schizophr. Res. 2014, 154, 42–47. [Google Scholar] [CrossRef] [PubMed]
  56. Rose, E.J.; Morris, D.W.; Hargreaves, A.; Fahey, C.; Greene, C.; Garavan, H.; Gill, M.; Corvin, A.; Donohoe, G. Neural effects of the CSMD 1 genome-wide associated schizophrenia risk variant rs10503253. Am. J. Med. Genet. Part B Neuropsychiatr. Genet. 2013, 162, 530–537. [Google Scholar] [CrossRef]
  57. Abd El Gayed, E.M.; Rizk, M.S.; Ramadan, A.N.; Bayomy, N.R. mRNA Expression of the CUB and Sushi Multiple Domains 1 (CSMD1) and Its Serum Protein Level as Predictors for Psychosis in the Familial High-Risk Children and Young Adults. ACS Omega 2021, 6, 24128–24138. [Google Scholar] [CrossRef]
  58. Ruiz-Martínez, J.; Azcona, L.J.; Bergareche, A.; Martí-Massó, J.F.; Paisán-Ruiz, C. Whole-exome sequencing associates novel CSMD1 gene mutations with familial Parkinson disease. Neurol. Genet. 2017, 3, e177. [Google Scholar] [CrossRef] [Green Version]
  59. Shahmohammadibeni, N.; Rahimi-Aliabadi, S.; Jamshidi, J.; Emamalizadeh, B.; Shahmohammadibeni, H.A.; Zare Bidoki, A.; Akhavan-Niaki, H.; Eftekhari, H.; Abdollahi, S.; Shekari Khaniani, M. The analysis of association between SNCA, HUSEYO and CSMD1 gene variants and Parkinson’s disease in Iranian population. Neurol. Sci. 2016, 37, 731–736. [Google Scholar] [CrossRef]
  60. Mitelman, F.; Mertens, F.; Johansson, B. A breakpoint map of recurrent chromosomal rearrangements in human neoplasia. Nat. Genet. 1997, 15, 417–474. [Google Scholar] [CrossRef]
  61. Sunwoo, J.B.; Holt, M.S.; Radford, D.M.; Deeker, C.; Scholnick, S.B. Evidence for multiple tumor suppressor genes on chromosome arm 8p in supraglottic laryngeal cancer. Genes Chromosomes Cancer 1996, 16, 164–169. [Google Scholar] [CrossRef]
  62. Wu, C.L.; Roz, L.; Sloan, P.; Read, A.P.; Holland, S.; Porter, S.; Scully, C.; Speight, P.M.; Thakker, N. Deletion mapping defines three discrete areas of allelic imbalance on chromosome arm 8p in oral and oropharyngeal squamous cell carcinomas. Genes Chromosomes Cancer 1997, 20, 347–353. [Google Scholar] [CrossRef]
  63. Ishwad, C.S.; Shuster, M.; Bockmühl, U.; Thakker, N.; Shah, P.; Toomes, C.; Dixon, M.; Ferrell, R.E.; Gollin, S.M. Frequent allelic loss and homozygous deletion in chromosome band 8p23 in oral cancer. Int. J. Cancer 1999, 80, 25–31. [Google Scholar] [CrossRef]
  64. Zhu, Q.; Gong, L.; Wang, J.; Tu, Q.; Yao, L.; Zhang, J.-R.; Han, X.-J.; Zhu, S.-J.; Wang, S.-M.; Li, Y.-H. miR-10b exerts oncogenic activity in human hepatocellular carcinoma cells by targeting expression of CUB and sushi multiple domains 1 (CSMD1). BMC Cancer 2016, 16, 806. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Bartha, Á.; Győrffy, B. TNMplot.com: A Web Tool for the Comparison of Gene Expression in Normal, Tumor and Metastatic Tissues. Int. J. Mol. Sci. 2021, 22, 2622. [Google Scholar] [CrossRef] [PubMed]
  66. Kamal, M.; Shaaban, A.M.; Zhang, L.; Walker, C.; Gray, S.; Thakker, N.; Toomes, C.; Speirs, V.; Bell, S.M. Loss of CSMD1 expression is associated with high tumour grade and poor survival in invasive ductal breast carcinoma. Breast Cancer Res. Treat. 2010, 121, 555–563. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Escudero-Esparza, A.; Bartoschek, M.; Gialeli, C.; Okroj, M.; Owen, S.; Jirström, K.; Orimo, A.; Jiang, W.G.; Pietras, K.; Blom, A.M. Complement inhibitor CSMD1 acts as tumor suppressor in human breast cancer. Oncotarget 2016, 7, 76920. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Deng, N.; Minoche, A.; Harvey, K.; Li, M.; Winkler, J.; Goga, A.; Swarbrick, A. Deep whole genome sequencing identifies recurrent genomic alterations in commonly used breast cancer cell lines and patient-derived xenograft models. Breast Cancer Res. 2022, 24, 63. [Google Scholar] [CrossRef]
  69. Gialeli, C.; Tuysuz, E.C.; Staaf, J.; Guleed, S.; Paciorek, V.; Mörgelin, M.; Papadakos, K.S.; Blom, A.M. Complement inhibitor CSMD1 modulates epidermal growth factor receptor oncogenic signaling and sensitizes breast cancer cells to chemotherapy. J. Exp. Clin. Cancer Res. 2021, 40, 258. [Google Scholar] [CrossRef]
  70. Paris, P.L.; Andaya, A.; Fridlyand, J.; Jain, A.N.; Weinberg, V.; Kowbel, D.; Brebner, J.H.; Simko, J.; Watson, J.V.; Volik, S. Whole genome scanning identifies genotypes associated with recurrence and metastasis in prostate tumors. Hum. Mol. Genet. 2004, 13, 1303–1313. [Google Scholar] [CrossRef]
  71. Fan, X.; Song, J.; Fan, Y.; Li, J.; Chen, Y.; Zhu, H.; Zhang, Z. CSMD1 Mutation Related to Immunity Can Be Used as a Marker to Evaluate the Clinical Therapeutic Effect and Prognosis of Patients with Esophageal Cancer. Int. J. Gen. Med. 2021, 14, 8689. [Google Scholar] [CrossRef]
  72. Chen, X.-L.; Hong, L.-L.; Wang, K.-L.; Liu, X.; Wang, J.-L.; Lei, L.; Xu, Z.-Y.; Cheng, X.-D.; Ling, Z.-Q. Deregulation of CSMD1 targeted by microRNA-10b drives gastric cancer progression through the NF-κB pathway. Int. J. Biol. Sci. 2019, 15, 2075. [Google Scholar] [CrossRef] [Green Version]
  73. Huang, T.; Liang, Y.; Zhang, H.; Chen, X.; Wei, H.; Sun, W.; Wang, Y. CSMD1 mutations are associated with increased mutational burden, favorable prognosis, and anti-tumor immunity in gastric cancer. Genes 2021, 12, 1715. [Google Scholar] [CrossRef] [PubMed]
  74. Wang, X.; Wang, S.; Han, Y.; Xu, M.; Li, P.; Ke, M.; Teng, Z.; Huang, P.; Diao, Z.; Yan, Y. Association of CSMD1 with Tumor Mutation Burden and Other Clinical Outcomes in Gastric Cancer. Int. J. Gen. Med. 2021, 14, 8293. [Google Scholar] [CrossRef]
  75. Liu, H.; Chen, Y.; Zhou, L.; Jiang, X.; Zhou, X. MicroRNA-642b-3p functions as an oncomiR in gastric cancer by down-regulating the CUB and sushi multiple domains protein 1/smad axis. Bioengineered 2022, 13, 9614–9628. [Google Scholar] [CrossRef] [PubMed]
  76. Liu, J.; Xu, R.; Mai, S.-J.; Ma, Y.-S.; Zhang, M.-Y.; Cao, P.-S.; Weng, N.-Q.; Wang, R.-Q.; Cao, D.; Wei, W. LncRNA CSMD1-1 promotes the progression of hepatocellular carcinoma by activating MYC signaling. Theranostics 2020, 10, 7527. [Google Scholar] [CrossRef] [PubMed]
  77. Zhao, Y.; Yang, B.; Chen, D.; Zhou, X.; Wang, M.; Jiang, J.; Wei, L.; Chen, Z. Combined identification of ARID1A, CSMD1, and SENP3 as effective prognostic biomarkers for hepatocellular carcinoma. Aging 2021, 13, 4696. [Google Scholar] [CrossRef] [PubMed]
  78. Fidler, I.J. The pathogenesis of cancer metastasis: The ‘seed and soil’ hypothesis revisited. Nat. Rev. Cancer 2003, 3, 453–458. [Google Scholar] [CrossRef]
  79. Qin, L.-X. Chromosomal aberrations related to metastasis of human solid tumors. World J. Gastroenterol. 2002, 8, 769. [Google Scholar] [CrossRef]
  80. Lang, M.-F.; Yang, S.; Zhao, C.; Sun, G.; Murai, K.; Wu, X.; Wang, J.; Gao, H.; Brown, C.E.; Liu, X. Genome-wide profiling identified a set of miRNAs that are differentially expressed in glioblastoma stem cells and normal neural stem cells. PLoS ONE 2012, 7, e36248. [Google Scholar] [CrossRef]
  81. Kwon, E.; Wang, W.; Tsai, L. Validation of schizophrenia-associated genes CSMD1, C10orf26, CACNA1C and TCF4 as miR-137 targets. Mol. Psychiatry 2013, 18, 11–12. [Google Scholar] [CrossRef]
  82. Wang, X.; Chen, X.; Liu, Y.; Huang, S.; Ding, J.; Wang, B.; Dong, P.; Sun, Z.; Chen, L. CSMD1 suppresses cancer progression by inhibiting proliferation, epithelial-mesenchymal transition, chemotherapy-resistance and inducing immunosuppression in esophageal squamous cell carcinoma. Exp. Cell Res. 2022, 417, 113220. [Google Scholar] [CrossRef]
Figure 1. Schematic illustration of the CUB and Sushi Multiple Domains 1 (CSMD1) gene and protein structures.
Figure 1. Schematic illustration of the CUB and Sushi Multiple Domains 1 (CSMD1) gene and protein structures.
Genes 13 02332 g001
Figure 2. Frequency and type of genetic alterations at the CSMD1 locus in different cancer types.
Figure 2. Frequency and type of genetic alterations at the CSMD1 locus in different cancer types.
Genes 13 02332 g002
Figure 3. Summary of CSMD1-mediated functions.
Figure 3. Summary of CSMD1-mediated functions.
Genes 13 02332 g003
Figure 4. Overview of complement system and CSMD1.
Figure 4. Overview of complement system and CSMD1.
Genes 13 02332 g004
Figure 5. CSMD1 differential gene expression in normal and tumour tissues. CSMD1 is overexpressed in a number of tumour types compared to normal tissue, with AML and liver being the highest. However, CSMD1 expression is not strongly observed in renal and prostate tumours. A Mann–Whitney U test was performed to mark the significant difference in expression between normal and tumour samples depicted by red colour.
Figure 5. CSMD1 differential gene expression in normal and tumour tissues. CSMD1 is overexpressed in a number of tumour types compared to normal tissue, with AML and liver being the highest. However, CSMD1 expression is not strongly observed in renal and prostate tumours. A Mann–Whitney U test was performed to mark the significant difference in expression between normal and tumour samples depicted by red colour.
Genes 13 02332 g005
Figure 6. Brief summary of EMT, MET, and CSMD1.
Figure 6. Brief summary of EMT, MET, and CSMD1.
Genes 13 02332 g006
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Ermis Akyuz, E.; Bell, S.M. The Diverse Role of CUB and Sushi Multiple Domains 1 (CSMD1) in Human Diseases. Genes 2022, 13, 2332. https://doi.org/10.3390/genes13122332

AMA Style

Ermis Akyuz E, Bell SM. The Diverse Role of CUB and Sushi Multiple Domains 1 (CSMD1) in Human Diseases. Genes. 2022; 13(12):2332. https://doi.org/10.3390/genes13122332

Chicago/Turabian Style

Ermis Akyuz, Esra, and Sandra M. Bell. 2022. "The Diverse Role of CUB and Sushi Multiple Domains 1 (CSMD1) in Human Diseases" Genes 13, no. 12: 2332. https://doi.org/10.3390/genes13122332

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop