bims-instec Biomed News
on Intestinal stem cells and chemoresistance in colon cancer and intestinal regeneration
Issue of 2022‒11‒20
34 papers selected by
Maria-Virginia Giolito
Free University of Brussels


  1. Nat Metab. 2022 Nov 17.
      The small intestine displays marked anatomical and functional plasticity that includes adaptive alterations in adult gut morphology, enteroendocrine cell profile and their hormone secretion, as well as nutrient utilization and storage. In this Perspective, we examine how shifts in dietary and environmental conditions bring about changes in gut size, and describe how the intestine adapts to changes in internal state, bowel resection and gastric bypass surgery. We highlight the critical importance of these intestinal remodelling processes in maintaining energy balance of the organism, and in protecting the metabolism of other organs. The intestinal resizing is supported by changes in the microbiota composition, and by activation of carbohydrate and fatty acid metabolism, which govern the intestinal stem cell proliferation, intestinal cell fate, as well as survivability of differentiated epithelial cells. The discovery that intestinal remodelling is part of the normal physiological adaptation to various triggers, and the potential for harnessing the reversible gut plasticity, in our view, holds extraordinary promise for developing therapeutic approaches against metabolic and inflammatory diseases.
    DOI:  https://doi.org/10.1038/s42255-022-00679-6
  2. Trends Cell Biol. 2022 Nov 14. pii: S0962-8924(22)00234-3. [Epub ahead of print]
      Proper regulation of ion balance across the intestinal epithelium is essential for physiological functions, while ion imbalance causes intestinal disorders with dire health consequences. Ion channels, pumps, and exchangers are vital for regulating ion movements (i.e., bioelectric currents) that control epithelial absorption and secretion. Recent in vivo studies used the Drosophila gut to identify conserved pathways that link regulators of Ca2+, Na+ and Cl- with intestinal stem cell (ISC) proliferation. These studies laid a foundation for using the Drosophila gut to identify conserved proliferative responses triggered by bioelectric regulators. Here, we review these studies, discuss their significance, as well as the advantages of using Drosophila to unravel conserved bioelectrically induced molecular pathways in the intestinal epithelium under physiological, pathophysiological, and regenerative conditions.
    Keywords:  Drosophila; bioelectric signaling; gut; intestinal stem cells; ion channels
    DOI:  https://doi.org/10.1016/j.tcb.2022.10.003
  3. Eur J Cell Biol. 2022 Nov 04. pii: S0171-9335(22)00085-1. [Epub ahead of print]101(4): 151282
      Calorie restriction has been recently shown to increase intestinal stem cell competition and to reduce mutation fixation in young mice. However, the impact of aging on this process is unknown. By employing Confetti reporter mice, here we show that, unexpectedly, old mice have more intestinal stem cell (ISC) competition than young mice. Moreover, differently from what observed in young mice, calorie restriction, when applied at late-life, decreases this process. Importantly, we also observed a strong correlation between the ISC competition and Paneth cell number. In vivo analysis and in vitro organoid experiments indicated that Paneth cells play a major role in driving intestinal stem cell competition and crypt clonality. Taken together, our results provide evidence that increasing the number of Paneth cells can increase the number of competitive ISCs, representing a valuable therapeutic target to delay fixation of mutated intestinal stem cells.
    Keywords:  Aging; Calorie restriction; Clonal drift; Intestinal stem cells; Niche paneth cells
    DOI:  https://doi.org/10.1016/j.ejcb.2022.151282
  4. J Gastrointest Oncol. 2022 Oct;13(5): 2458-2471
      Background: In recent years, reports regarding stimulator of interferon genes (STING) and the progression of colorectal cancer (CRC) have emerged rapidly, yet their association remains controversial. This research was aimed to provide an insight into the prognostic biomarker and therapeutic target significance of STING in CRC.Methods: CRC Cell lines of HCT116 and SW480, as well as 32 paired CRC specimens were chosen for this study. STING expressions were examined by immunohistochemistry to evaluate the correlation with clinicopathological factors. Data analysis of STING expressions in colon cancer and rectal cancer were performed using The Cancer Genome Atlas (TCGA) database. siRNA was transfected into cell lines for knocking down the expression of STING. Transwell assay was employed to evaluate cell migration and invasiveness. CCK-8 assay was used for assessing the change of cell proliferation. Drug sensitive test was involved to evaluate drug resistance of cell lines. Gene Set Enrichment Analysis (GSEA) was applied for exploring potential downstream mechanism of STING in CRC progression and Western blotting is used for mechanism validation.
    Results: In the thirty-two paired CRC and adjacent normal tissues, we found a significant up-regulated in STING expression with immunohistochemical staining in cancer tissues compared with adjacent normal tissues (P<0.01), which was correlated with the tumor-node-metastasis (TNM) stage of patients (P=0.028). Meanwhile, GESA enrichment analysis indicated a remarkable change in mTOR signaling following STING regulation. In HCT116 and SW480 cell lines of CRC, When STING was down-regulated, its biological behavior of cell viability, cell invasion and drug sensitivity to 5-fluorouracil were significantly reduced (P<0.05), we also observed the up-regulation of P-AMPK (P<0.05) and down-regulation of p-mTOR (P<0.05).
    Conclusions: STING expressions was significantly up-regulated in CRC tissues. Expression of STING was correlated with the TNM stage of patients. STING is found to promote cell proliferation, invasion ability and drug resistance mediating AMPK-mTOR signaling in CRC. STING could be a promising target for the sensitization of chemotherapy and inhibits CRC progression.
    Keywords:  Colorectal cancer (CRC); clinicopathological features; drug resistance; proliferation; stimulator of interferon genes (STING)
    DOI:  https://doi.org/10.21037/jgo-22-957
  5. J Pathol. 2022 Nov 14.
      Radiation enteritis (RE) is a prevalent complication of radiotherapy for pelvic malignant tumors, characterized by severe intestinal epithelial destruction and progressive submucosal fibrosis. However, little is known about the pathogenesis of this disease and so far, there is no specific targeted therapy. Here, we report that CXCL16 is up-regulated in the injured intestinal tissues of RE patients and in a mouse model. Genetic deletion of Cxcl16 mitigates fibrosis and promotes intestinal stem cell-mediated epithelial regeneration after radiation injury in mice. Mechanistically, CXCL16 functions on myofibroblasts through its receptor CXCR6 and activates JAK3/STAT3 signaling to promote fibrosis, and meanwhile to transcriptionally modulate the levels of BMP4 and HGF in myofibroblasts. Moreover, we find that CXCL16 and CXCR6 auto- and cross-regulate themselves in positive feedback loops. Treatment with CXCL16 neutralizing monoclonal antibody attenuates fibrosis and improves the epithelial repair in RE mouse model. Our findings emphasize the important role of CXCL16 in the progression of RE, and suggest that CXCL16 signaling could be a potential therapeutic target for RE. This article is protected by copyright. All rights reserved.
    Keywords:  BMP4; CXCL16; HGF; JAK3/STAT3 signaling; epithelial repair; fibrosis; intestinal stem cell; myofibroblast; radiation enteritis
    DOI:  https://doi.org/10.1002/path.6031
  6. Clin Transl Oncol. 2022 Nov 14.
      Different types of cells that are involved in tumor immunity play a significant part in antitumor therapy. The intestinal microbiota consist of the trillions of diverse microorganisms that inhabit the gastrointestinal tract. Recently, much emphasis has been paid to the link between these symbionts and colorectal cancer (CRC). This association might be anything from oncogenesis and cancer development to resistance or susceptibility to chemotherapeutic medicines. Cancer patients have a significantly different microbial composition in their guts compared to healthy persons. The microbiome may play a role in the development and development of cancer through the modulation of tumor immunosurveillance, as shown by these studies; however, the specific processes underlying this role are still poorly understood. This review focuses on the relationship between the intestinal bacterial microbiota and immune cells to determine how the commensal microbiome influences the initiation and development of CRC.
    Keywords:  Colorectal cancer; Dysbiosis; Immune cells; Microbiota
    DOI:  https://doi.org/10.1007/s12094-022-02995-5
  7. Transl Oncol. 2022 Nov 10. pii: S1936-5233(22)00229-7. [Epub ahead of print]27 101570
      BACKGROUND: The tumor microenvironment (TME) plays a critical role in shaping tumor progression and determining the outcome of the therapeutic response. In this study, we aimed to generate a comprehensive cellular landscape of the colorectal cancer (CRC) TME.METHODS: We generated a comprehensive single-cell atlas by collecting CRC cases that have been uploaded to the online database and conducting an in-depth secondary analysis. We then carried out spatial transcriptomic sequencing and multiple immunohistochemical analyses to verify the results of the single-cell analysis. Moreover, we applied our findings to the TCGA database and used tissue microarray (TMA) on CRC tissue specimens to validate clinical prognosis.
    FINDINGS: We re-analyzed the transcriptomes of 23785 cells, revealing a pattern of cell heterogeneity in the tumor region, leading-edge region, and non-tumor region. A subtype of COL11A1+INHBA+ tumor-resident cancer-associated fibroblasts (CAFs) was identified, and marker genes, transcription factors, and tissue-specific expression differences were noted and suggested to have potential roles in promoting cancer. We further confirmed that COL11A1+INHBA+ tumor-resident CAFs are mainly located in the hypoxic TME and we propose that they interact with CD44+ CRC cells via INHBA. Elevation of INHBA in CRC is associated with a poor prognosis.
    INTERPRETATION: Our results demonstrated a single cell landscape of CRC in different regions and identified in hypoxic TME a special subtype of CAFs producing INHBA, which promotes CRC development and correlates with poor prognosis. This special subtype of CAFs is a candidate target for translational research.
    Keywords:  Cancer-associated fibroblasts; Colorectal cancer; Hypoxia; Tumor microenvironment
    DOI:  https://doi.org/10.1016/j.tranon.2022.101570
  8. J Gastrointest Oncol. 2022 Oct;13(5): 2340-2350
      Background: Colorectal cancer (CRC) is a tumor with high incidence and poor prognosis. An increasing number of studies have shown that intermediate filament proteins, such as nestin, participate in the regulation of tumor progression. However, the mechanism related to CRC is complex, and the role and underlying mechanism of nestin have not been elucidated in CRC.Methods: We conducted quantitative reverse transcription polymerase chain reaction (RT-qPCR) and western blot analyses to examine the mRNA and protein levels in CRC and normal tissues. siRNAs targeting Nestin were transfected into CRC cells and then cell counting kit-8 (CCK-8), 5-ethynyl-2-deoxyuridine (EdU), sphere formation, and transwell analyses were used to assess the role of nestin in the proliferation, stem activity, migration, and invasive ability of CRC cells. Afterwards, nestin was overexpressed in CRC cells and P53 was overexpressed as a rescue group. CCK-8, EdU dyeing, sphere formation, and transwell assay was used to evaluated the role of Nestin/p53 axis in CRC cells.
    Results: We found high nestin expression and low p53 expression in CRC tissues and cells. Functionally, silencing of nestin suppressed the multiplication, stemness, and metastatic ability of Caco-2 and RKO cells. Encouragingly, rescue experiments suggested that overexpression of p53 partly restored the impacts of nestin overexpression on the viability, proliferation, and metastatic ability of CRC cells.
    Conclusions: We confirmed that nestin and p53 play a functional role in the progression of CRC, and they may act as potential therapeutic targets for CRC treatment.
    Keywords:  Nestin; colorectal cancer (CRC); p53; proliferation; stemness
    DOI:  https://doi.org/10.21037/jgo-22-788
  9. PLoS Comput Biol. 2022 Nov;18(11): e1010685
      5-Fluorouracil (5-FU) is a standard chemotherapeutic agent to treat solid cancers such as breast, colon, head, and neck. Computational modeling plays an essential role in predicting the outcome of chemotherapy and developing optimal dosing strategies. We developed an integrated mechanistic pharmacokinetics/pharmacodynamics (PK/PD) model examining the influence of 5-FU, as an S-phase specific double-strand break (DSB)-inducing agent, on tumor proliferation. The proposed mechanistic PK/PD model simulates the dynamics of critical intermediate components and provides the accurate tumor response prediction. The integrated model is composed of PK, cellular, and tumor growth inhibition (TGI) sub-models, quantitatively capturing the essential drug-related physiological processes. In the cellular model, thymidylate synthase (TS) inhibition, resultant deoxynucleoside triphosphate (dNTP) pool imbalance, and DSB induction are considered, as well as 5-FU incorporation into RNA and DNA. The amount of 5-FU anabolites and DSBs were modeled to drive the kinetics of the pharmacological tumor response. Model parameters were estimated by fitting to literature data. Our simulation results successfully describe the kinetics of the intermediates regulating the 5-FU cytotoxic events and the pattern of tumor suppression. The comprehensive model simulated the tumor volume change under various dose regimens, and its generalizability was attested by comparing it with literature data. The potential causes of the tumor resistance to 5-FU are also investigated through Monte Carlo analysis. The simulation of various dosage regimens helps quantify the relationship between treatment protocols and chemotherapy potency, which will lead to the development of efficacy optimization.
    DOI:  https://doi.org/10.1371/journal.pcbi.1010685
  10. Front Immunol. 2022 ;13 1014834
      Background: Transient receptor potential channels (TRPC) play critical regulatory functions in cancer occurrence and progression. However, knowledge on its role in colorectal cancer (CRC) is limited. In addition, neoadjuvant treatment and immune checkpoint inhibitors (ICIs) have increasing roles in CRC management, but not all patients benefit from them. In this study, a TRPC related signature (TRPCRS) was constructed for prognosis, tumor immune microenvironment (TIME), and treatment response of CRC.Methods: Data on CRC gene expression and clinical features were retrospectively collected from TCGA and GEO databases. Twenty-eight TRPC regulators (TRPCR) were retrieved using gene set enrichment analysis. Different TRPCR expression patterns were identified using non-negative matrix factorization for consensus clustering, and a TRPCRS was established using LASSO. The potential value of TRPCRS was assessed using functional enrichment analysis, tumor immune analysis, tumor somatic mutation analysis, and response to preoperative chemoradiotherapy or ICIs. Moreover, an external validation was conducted using rectal cancer samples that received preoperative chemoradiotherapy at Fujian Cancer Hospital (FJCH) via qRT-PCR.
    Results: Among 834 CRC samples in the TCGA and meta-GEO cohorts, two TRPCR expression patterns were identified, which were associated with various immune infiltrations. In addition, 266 intersected genes from 5564 differentially expressed genes (DEGs) between two TRPC subtypes, 4605 DEGs between tumor tissue and adjacent non-tumor tissue (all FDR< 0.05, adjusted P< 0.001), and 1329 prognostic related genes (P< 0.05) were identified to establish the TRPCRS, which was confirmed in the TCGA cohort, two cohorts from GEO, and one qRT-PCR cohort from FJCH. According to the current signature, the high-TRPC score group had higher expressions of PD-1, PD-L1, and CTLA4, lower TIDE score, and improved response to anti-PD-1 treatment with better predictive ability. Compared to the high-TRPC score group, the low-TRPC score group comprised an immunosuppressive phenotype with increased infiltration of neutrophils and activated MAPK signaling pathway, but was more sensitive to preoperative chemoradiotherapy and associated with improved prognosis.
    Conclusions: The current TRPCRS predicted the prognosis of CRC, evaluated the TIME in CRC, and anticipated the response to immune therapy and neoadjuvant treatment.
    Keywords:  colorectal cancer; immune checkpoint inhibitor; neoadjuvant treatment; prognosis; transient receptor potential channels
    DOI:  https://doi.org/10.3389/fimmu.2022.1014834
  11. Mucosal Immunol. 2022 Nov 15.
      Rapid development of the fetal and neonatal intestine is required to meet the growth requirements of early life and form a protective barrier against external insults encountered by the intestinal mucosa. The fetus receives nutrition via the placenta and is protected from harmful pathogens in utero, which leads to intestinal development in a relatively quiescent environment. Upon delivery, the intestinal mucosa is suddenly tasked with providing host defense and meeting nutritional demands. To serve these functions, an array of specialized epithelial cells develop from intestinal stem cells starting in utero and continuing postnatally. Intestinal disease results when these homeostatic processes are interrupted. For preterm neonates, the most common pathology resulting from epithelial barrier dysfunction is necrotizing enterocolitis (NEC). In this review, we discuss the normal development and function of the intestinal epithelium in early life as well as how disruption of these processes can lead to NEC.
    DOI:  https://doi.org/10.1038/s41385-022-00579-8
  12. Cell Rep. 2022 Nov 15. pii: S2211-1247(22)01512-1. [Epub ahead of print]41(7): 111641
      Long-term impacts of diet have been well studied; however, the immediate response of the intestinal epithelium to a change in nutrients remains poorly understood. We use physiological metrics and single-cell transcriptomics to interrogate the intestinal epithelial cell response to a high-fat diet (HFD). Within 1 day of HFD exposure, mice exhibit altered whole-body physiology and increased intestinal epithelial proliferation. Single-cell transcriptional analysis on day 1 reveals a cell-stress response in intestinal crypts and a shift toward fatty acid metabolism. By 3 days of HFD, computational trajectory analysis suggests an emergence of progenitors, with a transcriptional profile shifting from secretory populations toward enterocytes. Furthermore, enterocytes upregulate lipid absorption genes and show increased lipid absorption in vivo over 7 days of HFD. These findings demonstrate the rapid intestinal epithelial response to a dietary change and help illustrate the essential ability of animals to adapt to shifting nutritional environments.
    Keywords:  CP: Cell biology; CP: Metabolism; enteroendocrine; fatty acid metabolism; high-fat diet; immediate dietary change; intestinal stem cells; lipid absorption
    DOI:  https://doi.org/10.1016/j.celrep.2022.111641
  13. Front Immunol. 2022 ;13 1040256
      More than 85% of colorectal cancer (CRC) patients, who are with microsatellite stability (MSS), are resistant to immune checkpoint blockade (ICB) treatment. To overcome this resistance, combination therapy with chemotherapy is the most common choice. However, many CRC patients do not benefit more from combination therapy than chemotherapy alone. We hypothesize that severe immunosuppression, caused by chemotherapy administered at the maximum tolerated dose, antagonizes the ICB treatment. In this study, we found that low-dose oxaliplatin (OX), an immunogenic cell death (ICD)-induced drug, increased the antitumor response of TIGIT blockade against CT26 tumor, which is regarded as a MSS tumor. Combined treatment with OX and TIGIT blockade fostered CD8+ T-cell infiltration into tumors and delayed tumor progression. Importantly, only low-dose immunogenic chemotherapeutics successfully sensitized CT26 tumors to TIGIT blockade. In contrast, full-dose OX induces severe immunosuppression and impaired the efficacy of combination therapy. Further, we also found that lack of synergy between nonimmunogenic chemotherapeutics and TIGIT blockade. Consequently, this study suggests that the strategies of combination treatment of chemotherapy and ICB should be re-evaluated. The chemotherapeutics should be chosen for the potential to ICD and the dosage and regimen should be also optimized.
    Keywords:  TIGIT blockade; colon cancer; immunogenic cell death; immunogenic chemotherapeutics; microsatellite stability
    DOI:  https://doi.org/10.3389/fimmu.2022.1040256
  14. J Cancer Res Clin Oncol. 2022 Nov 19.
      BACKGROUND: In metastatic colorectal cancer (mCRC), acquired resistance against anti-EGFR targeted monoclonal antibodies, such as cetuximab (CET), was shown to be frequently caused by activating alterations in the RAS genes KRAS or NRAS. To this day, no efficient follow-up treatment option has emerged to treat mCRC in such a setting of resistance.METHODS: To uncover potential targets for second-line targeted therapies, we used mass-spectrometric proteomics to shed light on kinome reprogramming in an established cellular model of acquired, KRAS-associated CET resistance.
    RESULTS: This CET resistance was reflected by significant changes in the kinome, most of them individual to each cell line. Interestingly, all investigated resistant cell lines displayed upregulation of the Ephrin type-A receptor 2 (EPHA2), a well-known driver of traits of progression. Expectedly resistant cell lines displayed increased migration (p < 0.01) that was significantly reduced by targeting the EPHA2 signalling axis using RNA interference (RNAi) (p < 0.001), ephrin-A1 stimulation (p < 0.001), dasatinib (p < 0.01), or anti-EPHA2 antibody treatment (p < 0.001), identifying it as an actionable target in mCRC with acquired CET resistance.
    CONCLUSION: These results highlight EPHA2 and its role in mCRC with KRAS-gene mutated acquired CET resistance and support its use as a potential actionable target for the development of future precision medicine therapies.
    Keywords:  Cetuximab resistance; Colorectal cancer; EPHA2; Molecular oncology; Proteomics
    DOI:  https://doi.org/10.1007/s00432-022-04416-0
  15. Mutat Res Rev Mutat Res. 2022 Nov 09. pii: S1383-5742(22)00035-7. [Epub ahead of print] 108445
      Colorectal cancer (CRC) arises by a continuous process of genetic diversification and clonal evolution. Multiple genes and pathways have a role in tumor initiation and progression. The gradual accumulation of genetic and epigenetic processes leads to the establishment of adenoma and cancer. The important 'driver' mutations in tumor suppressor genes (such as TP53, APC, and SMAD4) and oncogenes (such as KRAS, NRAS, MET, and PIK3CA) confer selective growth advantages and cause CRC advancement. Clonal evolution induced by therapeutic pressure, as well as intra-tumoral heterogeneity, has been a great challenge in the treatment of metastatic CRC. Tumors often develop resistance to treatments as a result of intra-tumor heterogeneity, clonal evolution, and selection. Hence, the development of a multidrug personalized approach should be prioritized to pave the way for therapeutics repurposing and combination therapy to arrest tumor progression. This review summarizes how selective drug pressure can impact tumor evolution, resulting in the formation of polyclonal resistance mechanisms, ultimately promoting cancer progression. Current strategies for targeting clonal evolution are described. By understanding sources and consequences of tumor heterogeneity, customized and effective treatment plans to combat drug resistance may be devised.
    Keywords:  Clonal evolution; Colorectal cancer (CRC); Metastasis; Subclonal mutations; Tumor heterogeneity
    DOI:  https://doi.org/10.1016/j.mrrev.2022.108445
  16. J Cell Mol Med. 2022 Nov 16.
      Platinum-based chemotherapy drugs play a very important role in the treatment of patients with advanced colorectal cancer, but the drug resistance of platinum-based chemotherapy drugs is an important topic that puzzles us. If we can find mechanisms of resistance, it will be revolutionary for us. We analysed the differential genes, core genes and their enrichment pathways in platinum-resistant and non-resistant patients through a public database. Platinum-resistant cell lines were cultured in vitro for in vitro colony and Transwell analysis. Tumorigenesis analysis of nude mice in vivo. Verify the function of core genes. Through differential gene and enrichment analysis, we found that CUL4B was the main factor affecting platinum drug resistance and EMT. Our hypothesis was further verified by in vitro drug-resistant and wild-type cell lines and in vivo tumorigenesis analysis of nude mice. CUL4B leads to platinum drug resistance in colorectal cancer by affecting tumour EMT.
    Keywords:  CUL4B; EMT; colorectal cancer; drug resistance
    DOI:  https://doi.org/10.1111/jcmm.17585
  17. Front Oncol. 2022 ;12 1018767
      The majority of colorectal cancers (CRCs) are microsatellite stable (MSS) and resistant to immunotherapy. The current study explores the possibility of using oncolytic reovirus to sensitize MSS CRC to immune checkpoint inhibition. While reovirus reduced metabolic activity among KRAS Mut cells, microarray/computational analysis revealed microsatellite status-oriented activation of immune-response pathways. Reovirus plus anti-PD-1 treatment increased cell death among MSS cells ex vivo. Reduced tumorigenicity and proliferative index, and increased apoptosis were evident among CT26 [MSS, KRAS Mut], but not in MC38 [microsatellite unstable/MSI, KRAS Wt] syngeneic mouse models under combinatorial treatment. PD-L1-PD-1 signaling axis were differentially altered among CT26/MC38 models. Combinatorial treatment activated the innate immune system, pattern recognition receptors, and antigen presentation markers. Furthermore, we observed the reduction of immunosuppressive macrophages and expansion of effector T cell subsets, as well as reduction in T cell exhaustion. The current investigation sheds light on the immunological mechanisms of the reovirus-anti-PD-1 combination to reduce the growth of MSS CRC.
    Keywords:  anti-PD-1; colorectal cancer; combinatorial therapy; immune checkpoint; microsatellite instability; reovirus; translational
    DOI:  https://doi.org/10.3389/fonc.2022.1018767
  18. Oncogene. 2022 Nov 14.
      The microenvironment of distant organ plays vital roles in regulating tumor metastases. However, little is known about the crosstalk between metastasized tumor cells and target organs. Herein, we found that EFNB2 expression was upregulated in liver metastases (LM) of colorectal cancer (CRC), but not in pulmonary metastases (PM) or primary CRC tumors. EFNB2 played a tumor-promoting role in CRC LM in vitro and in vivo. Through forward signaling, EFNB2-promoted CRC LM by interacting with the EPHB4 receptor. EFNB2/EPHB4 axis enhances LDLR-mediated cholesterol uptake in CRC LM. Subsequently, EFNB2/EPHB4 axis promotes LDLR transcription by regulating STAT3 phosphorylation. Blocking LDLR reversed the role of the EFNB2/EPHB4 axis in promoting CRC LM. Using clinical data, survival analysis revealed that the survival time of patients with CRC LM was decreased in patients with high EFNB2 expression, compared with low EFNB2 expression. Inhibition of the EFNB2/EPHB4 axis markedly prolonged the survival time of BALB/c nude mice with CRC LM with a high cholesterol diet. These findings revealed a key step in the regulation of cholesterol uptake by EFNB2/EPHB4 axis and its tumor-promoting role in CRC LM.
    DOI:  https://doi.org/10.1038/s41388-022-02519-z
  19. Proc Natl Acad Sci U S A. 2022 Nov 08. 119(45): e2205110119
      During coordinated development of two neighboring organs from the same germ layer, how precursors of one organ resist the inductive signals of the other to avoid being misinduced to wrong cell fate remains a general question in developmental biology. The liver and anterior intestinal precursors located in close proximity along the gut axis represent a typical example. Here we identify a zebrafish leberwurst (lbw) mutant with a unique hepatized intestine phenotype, exhibiting replacement of anterior intestinal cells by liver cells. lbw encodes the Cdx1b homeoprotein, which is specifically expressed in the intestine, and its precursor cells. Mechanistically, in the intestinal precursors, Cdx1b binds to genomic DNA at the regulatory region of secreted frizzled related protein 5 (sfrp5) to activate sfrp5 transcription. Sfrp5 blocks the mesoderm-derived, liver-inductive Wnt2bb signal, thus conferring intestinal precursor cells resistance to Wnt2bb. These results demonstrate that the intestinal precursors avoid being misinduced toward hepatic lineages through the activation of the Cdx1b-Sfrp5 cascade, implicating Cdx/Sfrp5 as a potential pharmacological target for the manipulation of intestinal-hepatic bifurcations, and shedding light on the general question of how precursor cells resist incorrect inductive signals during embryonic development.
    Keywords:  Cdx; Wnt; cell fate; intestine; liver
    DOI:  https://doi.org/10.1073/pnas.2205110119
  20. Am J Cancer Res. 2022 ;12(10): 4751-4763
      Bromodomain Adjacent to Zinc Finger Domain 1B (BAZ1B) is involved in multiple nuclear processes, and its role in tumorigenesis is emerging. However, the function of BAZ1B in colorectal cancer (CRC) remains largely unexplored. High-density tissue microarrays comprising 100 pairs of matched normal colon and treatment-naïve CRC samples were analyzed by immunohistochemistry with an anti-BAZ1B antibody. The HCT116 and SW480 CRC cell lines were used for overexpression and small hairpin RNA-mediated BAZ1B knockdown models, respectively. Both cell lines were xenografted to immunodeficient NU/J mice to assess tumor burden. The molecular consequences of alterations of BAZ1B expression were assessed by RNA-Seq of xenografts and functional analyses using the Reactome database. Immunohistochemical analysis of BAZ1B showed that BAZ1B staining intensity was higher in 93 tumor specimens and significantly correlated with tumor size (P = 0.03), but not with the presence of KRAS mutation. BAZ1B overexpression significantly increased and its knockdown inhibited the proliferation of HCT116 and SW480 cell lines, respectively. These findings were reproduced when both cell lines were grown as xenografts. RNA-Seq of HCT116 and SW480 xenografts identified 2046 and 99 differentially expressed genes (DEGs) (adjusted P ≤ 0.05), respectively. Functional annotation of DEGs identified already established as well as new molecular processes dependent on BAZ1B protein expression. In conclusion, BAZ1B is overexpressed in CRC tissue and contributes to CRC cell proliferation in vitro and in vivo. The data support the emerging oncogenic role of BAZ1B in cancerogenesis including in CRC.
    Keywords:  BAZ1B; RNA-Seq; WSTF; colorectal cancer; oncogene; xenografts
  21. Front Oncol. 2022 ;12 986103
      Women with colorectal cancer (CRC) have survival advantages over men, yet the underlying mechanisms are unclear. T cell infiltration within the CRC tumor microenvironment (TME) correlates strongly with survival. We hypothesized that women with CRC have increased T cell infiltration and differential gene expression in the TME compared to men. Tissue microarrays comprising primary tumor, tumor infiltrated lymph nodes, and uninvolved colon were created from CRC patients. Proportions of CD4 positive (CD4+) and CD8 positive (CD8+) T cells were identified using immunohistochemistry. TME immune- and cancer-related genetic expression from primary and metastatic CRC tumor were also evaluated via the NanoStringIO360 panel and The Cancer Genome Atlas Project database. CD4+ was higher in tumor samples from women compared to men (22.04% vs. 10.26%, p=0.002) and also in lymph node samples (39.54% vs. 8.56%, p=0.001). CD8+ was increased in uninvolved colon from women compared to men (59.40% vs. 43.61%, p=0.015), and in stage I/II tumors compared to III/IV in all patients (37.01% vs. 23.91%, p=0.009). Top CD8+ tertile patients survived longer compared to the bottom (43.9 months vs. 25.3 months, p=0.007). Differential gene expression was observed in pathways related to Treg function, T cell activity, and T cell exhaustion, amongst several others, in women compared to men. Thus, significant sexual dimorphism exists in the TME that could contribute to survival advantages observed in female patients with CRC.
    Keywords:  T cell; colorectal cancer; immunoscore; sexual dimorphism; tumor microenvironment
    DOI:  https://doi.org/10.3389/fonc.2022.986103
  22. Clin Chim Acta. 2022 Nov 11. pii: S0009-8981(22)01367-5. [Epub ahead of print]538 53-59
      BACKGROUND AND AIMS: Systemic chemotherapy followed by hepatic resection is the treatment of choice for patients with colorectal cancer liver metastasis (CRCLM) but reliable biomarkers predicting response to therapy are needed. Spontaneous apoptosis of single tumour cells is common in CRCLM. We explored the potential of circulating apoptosis markers to predict treatment response.MATERIALS AND METHODS: Fifty-eight patients with CRCLM or hepatocellular carcinoma (HCC) were included in this study. Tumour tissue and blood samples were obtained before and after initiation of chemotherapy. Immunohistochemistry and ELISA assays were utilized to quantify the apoptosis marker caspase-cleaved cytokeratin 18 (M30) in tissue and circulation.
    RESULTS: CRCLM tissues showed more apoptotic tumour cells than HCC, or healthy liver. This was associated with elevated levels of circulating M30 (median = 244 U/l vs. 37 U/l in healthy controls, p = 0.009) which correlated with tumour volume (r2 = 0.92). Patients with progressive disease during chemotherapy showed higher M30 levels before therapy than responders (745 U/l vs. 136 U/l, p = 0.016). The predictive potential of M30 was higher than that of the tumour markers CA19-9 or CEA (AUC: 0.93, 0.63, and 0.78, respectively).
    CONCLUSIONS: Apoptotic tumour cells release cellular debris into the circulation, which provides information about tumour size and vitality.
    Keywords:  Apoptosis; Caspase-cleaved cytokeratin 18; Colorectal cancer liver metastasis
    DOI:  https://doi.org/10.1016/j.cca.2022.11.009
  23. Front Oncol. 2022 ;12 898966
      Colorectal diseases are threatening human health, especially inflammatory bowel disease (IBD) and colorectal cancer (CRC). IBD is a group of chronic, recurrent and incurable disease, which may affect the entire gastrointestinal tract, increasing the risk of CRC. Eukaryotic gene expression is a complicated process, which is mainly regulated at the level of gene transcription and mRNA translation. Protein translation in tissue is associated with a sequence of steps, including initiation, elongation, termination and recycling. Abnormal regulation of gene expression is the key to the pathogenesis of CRC. In the early stages of cancer, it is vital to identify new diagnostic and therapeutic targets and biomarkers. This review presented current knowledge on aberrant expression of eIFs, eEFs and eRFs in colorectal diseases. The current findings of protein synthesis on colorectal pathogenesis showed that eIFs, eEFs and eRFs may be potential targets for CRC treatment.
    Keywords:  colorectal cancer; colorectal pathogenesis; eukaryotic gene expression; inflammatory bowel disease; protein translation
    DOI:  https://doi.org/10.3389/fonc.2022.898966
  24. J Gastrointest Oncol. 2022 Oct;13(5): 2426-2438
      Background: Colorectal cancer (CRC) is a common global malignancy associated with high invasiveness, high metastasis, and poor prognosis. CRC commonly metastasizes to the liver, where the treatment of metastasis is both difficult and an important topic in current CRC management.Methods: Microarrays data of human CRC with liver metastasis (CRCLM) were downloaded from the National Center for Biotechnology Information (NCBI) Gene Expression Omnibus (GEO) database to identify potential key genes. Differentially expressed (DE) genes (DEGs) and DEmiRNAs of primary CRC tumor tissues and metastatic liver tissues were identified. Microenvironment Cell Populations (MCP)-counter was used to estimate the abundance of immune cells in the tumor micro-environment (TME), and weighted gene correlation network analysis (WGCNA) was used to construct the co-expression network analysis. Gene Ontology and Kyoto Encyclopaedia of Gene and Genome (KEGG) pathway enrichment analyses were conducted, and the protein-protein interaction (PPI) network for the DEGs were constructed and gene modules were screened.
    Results: Thirty-five pairs of matched colorectal primary cancer and liver metastatic gene expression profiles were screened, and 610 DEGs (265 up-regulated and 345 down-regulated) and 284 DEmiRNAs were identified. The DEGs were mainly enriched in the complement and coagulation cascade pathways and renin secretion. Immune infiltrating cells including neutrophils, monocytic lineage, and cancer-associated fibroblasts (CAFs) differed significantly between primary tumor tissues and metastatic liver tissues. WGCN analysis obtained 12 modules and identified 62 genes with significant interactions which were mainly related to complement and coagulation cascade and the focal adhesion pathway. The best subset regression analysis and backward stepwise regression analysis were performed, and eight genes were determined, including F10, FGG, KNG1, MBL2, PROC, SERPINA1, CAV1, and SPP1. Further analysis showed four genes, including FGG, KNG1, CAV1, and SPP1 were significantly associated with CRCLM.
    Conclusions: Our study implies complement and coagulation cascade and the focal adhesion pathway play a significant role in the development and progression of CRCLM, and FGG, KNG1, CAV1, and SPP1 may be metastatic markers for its early diagnosis.
    Keywords:  Colorectal liver metastasis; co-expression network; complement system activation; metastasis markers; tumor immune microenvironments
    DOI:  https://doi.org/10.21037/jgo-22-965
  25. J Gastrointest Oncol. 2022 Oct;13(5): 2439-2446
      Background: Colorectal cancer (CRC) is one of the most common malignancies. Although CRC treatment has been significantly improved, patient survival remains low because most patients already have advanced disease at diagnosis. Early screening and diagnosis of tumors is critical; however, the current tissue biopsy and radiological evaluation methods have very limited effectiveness. Therefore, establishing new convenient and non-invasive biomarkers is urgently needed for timely detection, therapeutic assessment, and prognostic prediction. At present, non-coding RNAs (ncRNAs) have attracted research attention owing to their potential oncological applications.Methods: The long ncRNA epidermal growth factor receptor antisense RNA 1 (EGFR-AS1) is overexpressed in multiple malignancies including CRC. The present study examined the circulating EGFR-AS1 level in CRC, and the results showed that EGFR-AS1 could be considered an indicator of tumor burden.
    Results: Elevated circulating EGFR-AS1 levels were detected in CRC cases (n=128) compared with control cases comprising endoscopy confirmed CRC-free individuals [n=64, median expression normalized to glyceraldehyde-3-phosphate dehydrogenase (GAPDH), 1.578 vs. 0.780, P<0.001]. Individuals with larger tumors (≥5 cm) had elevated circulating EGFR-AS1 levels compared to those with smaller tumors (<5 cm, 1.739 vs. 1.290, P<0.001). The expression of serum EGFR-AS1 in stage III/IV CRC was higher than that in stage I/II CRC (1.691 vs. 1.412, P<0.05). Plasma EGFR-AS1 levels were markedly reduced following surgical resection of colorectal lesions in a subset of patients [n=32, 1.192 (pre-surgery) vs. 0.692, P<0.001]. Furthermore, the expression of EGFR-AS1 in resected CRC tissues was significantly higher than that in paracancerous tissues (n=32, 1.336 vs. 0.487, P<0.001).
    Conclusions: These results highlight the potential of EGFR-AS1 as a diagnostic biomarker in CRC.
    Keywords:  Biomarker; circulating; colorectal cancer (CRC); diagnostic; epidermal growth factor receptor antisense RNA 1 (EGFR-AS1)
    DOI:  https://doi.org/10.21037/jgo-22-968
  26. Transl Cancer Res. 2022 Oct;11(10): 3686-3697
      Background: Colorectal cancer (CRC) is a common malignant tumor leading to poor prognosis and high mortality. Mannosidase alpha class 2A member 1 (MAN2A1) turns to oncogene through fusing Fer tyrosine kinase (FER) and associates with multiple cancer occurrence. In order to determine whether MAN2A1 can promote tumorigenesis and metastasis in CRC, we conducted a series of studies.Methods: We obtained gene expression and clinical data of CRC from The Cancer Genome Atlas (TCGA) databases. RNA raw counts data was merged by Python. Batch processing of univariate Cox regression analysis was performed to preliminary identify the genes associated with prognosis. Differentially expressed genes (DEGs) between lymph node metastasis (LNM) patients and non-LNM patients were identified via edgR in Sangerbox tool. Protein-protein interactive (PPI) network was constructed using Search Tool for the Retrieval of Interacting Genes (STRING) and Cytoscape software. Kaplan-Meier (KM) survival of CRC patients was analyzed by Sangerbox tool. Clinicopathologic characteristics of CRC patients were analyzed by SPSS statistics software. Differences in RNA expression levels of genes were validated in our cohort by real-time polymerase chain reaction (RT-PCR). Analyses of Signaling pathways and gene ontology were explored by gene set enrichment analysis (GSEA).
    Results: We first obtained 4,455 genes associated with the prognosis of CRC, and 998 of these genes were also DEGs in CRC between metastatic CRC tissues and in situ tissues. Therein, MAN2A1 expression was downregulated in LNM CRC compared with CRC in situ, also downregulated in CRC compared with adjacent normal tissues, and high gene expression levels of MAN2A1 was associated with better survival.
    Conclusions: Our study suggested that MAN2A1 could be a potential biomarker significantly related to prognosis and LNM of CRC patients.
    Keywords:  Colorectal cancer (CRC); bioinformatics analysis; biomarker
    DOI:  https://doi.org/10.21037/tcr-22-629
  27. Expert Rev Anticancer Ther. 2022 Nov 14.
      OBJECTIVE: : It remains controversial which targeted monoclonal antibodies combined with chemotherapy can provide better efficacy in wild-type KRAS/RAS metastatic colorectal cancer (mCRC) patients. Therefore, we used this meta-analysis to assess the latest evidence of clinical outcomes.MATERIALS AND METHODS: : We systematically searched PubMed, Web of Science, Cochrane Library and Embase databases for eligible studies published from database inception to May 2022. RevMan 5.4 was used to conduct the meta-analysis.
    RESULTS: : 11 RCTs involving a total of 3575 patients were included. Meta-analysis showed that EGFR inhibitors significantly prolonged the overall survival (OS) [HR = 0.83, 95%CI (0.73, 0.94), P = 0.003] and overall response rate (ORR) [RR = 1.11, 95%CI (1.05, 1.18), P = 0.0003] compared to VEGF inhibitors in wild-type KRAS/RAS mCRC patients, but no significant difference in progression-free survival (PFS) [HR = 0.96, 95%CI (0.87, 1.07), P = 0.50]. In subgroup analysis, the survival benefit of EGFR inhibitors was limited to first-line treatment.
    CONCLUSION: : ​Our study showed that EGFR inhibitors were superior to VEGF inhibitors in wild-type KRAS/RAS mCRC patients, especially in patients with first-line treatment. However, subsequent large sample, multi-center RCTs are needed to further verify our conclusions.
    Keywords:  Bevacizumab; Cetuximab; Chemotherapy; Meta analysis; Metastatic colorectal cancer; Panitumumab
    DOI:  https://doi.org/10.1080/14737140.2022.2147512
  28. Sci Rep. 2022 Nov 12. 12(1): 19359
      Colorectal cancer (CRC) is one of the most commonly diagnosed cancer types worldwide. Despite significant advances in prevention and diagnosis, CRC is still one of the leading causes of cancer-related mortality globally. RAB27A, the member of RAB27 family of small GTPases, is the critical protein for intracellular secretion and has been reported to promote tumor progression. However, it is controversial for the role of RAB27A in CRC progression, so we explored the exact function of RAB27A in CRC development in this study. Based on the stable colon cancer cell lines of RAB27A knockdown and ectopic expression, we found that RAB27A knockdown inhibited proliferation and clone formation of SW480 colon cancer cells, whereas ectopic expression of RAB27A in RKO colon cancer cells facilitated cell proliferation and clone formation, indicating that RAB27A is critical for colon cancer cell growth. In addition, our data demonstrated that the migration and invasion of colon cancer cells were suppressed by RAB27A knockdown, but promoted by RAB27A ectopic expression. Therefore, RAB27A is identified as an onco-protein in mediating CRC development, which may be a valuable prognostic indicator and potential therapeutic target for CRC.
    DOI:  https://doi.org/10.1038/s41598-022-23696-7
  29. J Gastrointest Oncol. 2022 Oct;13(5): 2351-2365
      Background: Colorectal cancer (CRC) is one of the most prominent malignant diseases, with a high incidence and a dismal prognosis. Metastasis to the liver is the leading cause of death in CRC patients. This study aimed to identify accurate metastatic biomarkers of CRC and investigate the potential molecular mechanisms of liver metastasis of colorectal cancer (LMCRC).Methods: Three independent datasets were screened and downloaded from the Gene Expression Omnibus (GEO) database. The GEO2R tool was used to identify differentially expressed genes (DEGs) in CRC tissues and liver metastases. Next, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were conducted using the Database for Annotation, Visualization, and Integrated Discovery (DAVID). Furthermore, the protein-protein interactions (PPIs) of the DEGs were analyzed using the Search Tool for the Retrieval of Interacting Genes (STRING) database, Cytoscape, and Molecular Complex Detection (MCODE). Next, the expression levels and Kaplan-Meier survival analysis of the target gene between normal colon and CRC tissues were performed by UALCAN. The expression of the target gene in tissues and cell lines was verified by quantitative reverse transcription-polymerase chain reaction (qRT-PCR), western blot, and immunohistochemistry (IHC) assay. The impact of the target gene on the proliferation, invasion, and migration ability of COAD cells was explored in vitro.
    Results: A total of 92 common DEGs were found in the three independent datasets. GO/KEGG enrichment analysis showed that the DEGs were mainly involved in 14 different pathways. The protein-protein interaction (PPI) network revealed that complement 5 (C5), the upstream gene of C8A in the complement system, was associated with C8 and other key hub genes. Meanwhile, the online UALCAN resource showed that C5 was up-regulated and facilitated malignant progression in COAD samples. Next, we confirmed that C5 remarkably increased and promoted cell proliferation, migration, and invasion in CRC cell lines, SW620 and SW480. The IHC assay showed C5 was also highly expressed in a majority of LMCRC tissues compared with paired CRC tissues.
    Conclusions: The findings of our integrated bioinformatics study suggest that complement C5 might serve as a potential therapeutic target in patients with CRC.
    Keywords:  Colorectal cancer (CRC); bioinformatics analysis; complement C5; liver metastasis of colorectal cancer (LMCRC)
    DOI:  https://doi.org/10.21037/jgo-22-829
  30. J Gastrointest Oncol. 2022 Oct;13(5): 2447-2457
      Background: The aim of this study was to investigate the inhibiting effect of transient receptor potential vanilloid 3 (TRPV3) on the proliferation and migration of colorectal cancer (CRC) cells and to explore the underlying mechanism.Methods: A microarray dataset from the publicly available Gene Expression Omnibus (GEO) database was used to investigate the prognostic value of TRPV3 in CRC. In addition, 100 CRC tissue samples were collected at our center to further validate its prognostic value at the protein level. Cell proliferation ability was detected by Cell Counting Kit-8 (CCK-8) assay, and cell migration ability was detected by transwell assay. Gene set variation analysis (GSVA) was performed to identify the potential pathways regulated by TRPV3.
    Results: Based on the largest microarray dataset (GSE39582), low expression of TRPV3 was found to be significantly associated with poor prognosis in CRC patients, and this result was successfully validated at our cancer center. Functional experiments showed that knockdown of TRPV3 enhanced cell proliferation and migration, while enforced TRPV3 expression exhibited the opposite effect. GSEA based on public microarray data revealed that the mitogen-activated protein kinase (MAPK) signaling pathway was notably activated in patients with low expression of TRPV3. Further experiments in vivo confirmed that TRPV3 silencing promoted cell proliferation and migration by activating the MAPK signaling pathway.
    Conclusions: Low expression of TRPV3, which stimulates cell proliferation and migration by provoking the MAPK signaling pathway, indicated poor prognosis in CRC patients.
    Keywords:  Colorectal cancer (CRC); cell proliferation and migration; mitogen-activated protein kinase signaling pathway (MAPK signaling pathway); transient receptor potential vanilloid 3 (TRPV3)
    DOI:  https://doi.org/10.21037/jgo-22-938
  31. R Soc Open Sci. 2022 Nov;9(11): 220186
      Epithelial-mesenchymal transition (EMT) and immunoevasion through upregulation of programmed death-ligand 1 (PD-L1) are important drivers of cancer progression. While EMT has been proposed to facilitate PD-L1-mediated immunosuppression, molecular mechanisms of their interaction remain obscure. Here, we provide insight into these mechanisms by proposing a mathematical model that describes the crosstalk between EMT and interferon gamma (IFNγ)-induced PD-L1 expression. Our model shows that via interaction with microRNA-200 (miR-200), the multi-stability of the EMT regulatory circuit is mirrored in PD-L1 levels, which are further amplified by IFNγ stimulation. This IFNγ-mediated effect is most prominent for cells in a fully mesenchymal state and less strong for those in an epithelial or partially mesenchymal state. In addition, bidirectional crosstalk between miR-200 and PD-L1 implies that IFNγ stimulation allows cells to undergo EMT for lower amounts of inducing signal, and the presence of IFNγ accelerates EMT and decelerates mesenchymal-epithelial transition (MET). Overall, our model agrees with published findings and provides insight into possible mechanisms behind EMT-mediated immune evasion, and primary, adaptive, or acquired resistance to immunotherapy. Our model can be used as a starting point to explore additional crosstalk mechanisms, as an improved understanding of these mechanisms is indispensable for developing better diagnostic and therapeutic options for cancer patients.
    Keywords:  PD-L1; epithelial-mesenchymal transition (EMT); immune evasion; interferon gamma
    DOI:  https://doi.org/10.1098/rsos.220186
  32. Cancer Sci. 2022 Nov 16.
      This trial was initiated to evaluate the efficacy and safety of pyrotinib in combination with trastuzumab in patients with HER2-positive recurrent / metastatic colorectal cancer (CRC). In this single-arm, open-label, muti-center, phase 2 trial, patients with HER2-positive recurrent / metastatic CRC were enrolled and received oral pyrotinib 400 mg once a day plus intravenous trastuzumab 8 mg/kg loading dose followed by 6 mg/kg once every 3 weeks. The primary endpoint was objective response rate (ORR). Disease control rate (DCR), progression-free survival (PFS), duration of response and safety were assessed as secondary endpoints. From December 2019 to October 2021, a total of 20 patients were enrolled and 18 of them were evaluable for response. All patients were BRAF wild-type. Four patients achieved partial response, with an ORR of 22.2% (4/18; 95% CI, 6.4 to 47.6) and DCR of 61.1% (11/18; 95% CI, 35.8 to 82.7). While the ORR and DCR were 33.3% (4/12; 95% CI, 13.8 to 60.9) and 83.3% (10/12; 95% CI, 51.6 to 97.9), respectively, in RAS wild-type patients. At the time of cut-off day, median follow-up was 10.7 months (range: 3.8 to 13.8). The median PFS was 3.4 months (95% CI, 1.8 to 4.3) in the overall population and 4.3 months (95% CI, 3.2 to 8.5) in the RAS wild-type group. The most common adverse event of grade ≥3 was diarrhea (13/20, 65.0%). Pyrotinib combined with trastuzumab showed promising antitumor activity and a manageable safety profile in patients with RAS/BRAF wild-type HER2-positive advanced CRC. ClinicalTrials.gov Identifier: NCT04380012.
    Keywords:  Pyrotinib; RAS mutation; colorectal cancer; human epidermal growth factor receptor 2 (HER2); trastuzumab
    DOI:  https://doi.org/10.1111/cas.15660
  33. Front Oncol. 2022 ;12 958155
      Human TRIAP1 (TP53-regulated inhibitor of apoptosis 1; also known as p53CSV for p53-inducible cell survival factor) is the homolog of yeast Mdm35, a well-known chaperone that interacts with the Ups/PRELI family proteins and participates in the intramitochondrial transfer of lipids for the synthesis of cardiolipin (CL) and phosphatidylethanolamine. Although recent reports indicate that TRIAP1 is a prosurvival factor abnormally overexpressed in various types of cancer, knowledge about its molecular and metabolic function in human cells is still elusive. It is therefore critical to understand the metabolic and proliferative advantages that TRIAP1 expression provides to cancer cells. Here, in a colorectal cancer cell model, we report that the expression of TRIAP1 supports cancer cell proliferation and tumorigenesis. Depletion of TRIAP1 perturbed the mitochondrial ultrastructure, without a major impact on CL levels and mitochondrial activity. TRIAP1 depletion caused extramitochondrial perturbations resulting in changes in the endoplasmic reticulum-dependent lipid homeostasis and induction of a p53-mediated stress response. Furthermore, we observed that TRIAP1 depletion conferred a robust p53-mediated resistance to the metabolic stress caused by glutamine deprivation. These findings highlight the importance of TRIAP1 in tumorigenesis and indicate that the loss of TRIAP1 has extramitochondrial consequences that could impact on the metabolic plasticity of cancer cells and their response to conditions of nutrient deprivation.
    Keywords:  TRIAP1/Mdm35; coiled-coil-helix-coiled-coil-helix domain (CHCHD)-containing proteins; colon cancer; glutamine starvation; lipid homeostasis; mitochondria; p53-mediated stress response
    DOI:  https://doi.org/10.3389/fonc.2022.958155
  34. Pathol Res Pract. 2022 Nov 05. pii: S0344-0338(22)00441-1. [Epub ahead of print]240 154197
      Hematological metastasis was the main metastatic method of colorectal cancer and the main reason for failure of radical surgery. Vascular endothelial cells played an important role in tumor hematologic metastasis. We previously performed RNA-Seq on primary and metastatic colorectal carcinoma (CRC) tissues and then identified GPR63 as a potential metastasis-promoting gene, but its role and mechanisms in the interaction between cancer cells and vascular endothelial cells were still unknown. In this study, GPR63 was significantly elevated in CRC tissues compared with paracarcinoma tissues. GPR63 expression was closely related to lymph node metastasis and distant metastasis in 147 CRC tissues. GPR63 promoted cell migration and stemness. Moreover, endothelial cell-derived S1P enhanced the migration and sphere-forming ability of CRC through activation of GPR63. Mechanistically, S1P promoted GPR63 binding to Src to activate JAK2/STAT3 pathway, and therefore promoted CRC cell migration. Our study revealed a novel mechanism by which endothelial cells promoted metastasis of CRC cells, which might have potential as a promising target for CRC therapy.
    Keywords:  CRC; Endothelial cell; GPR63; Migration; S1P
    DOI:  https://doi.org/10.1016/j.prp.2022.154197