Skip to main content

Alterations in complex lipids in tumor tissue of patients with colorectal cancer

Abstract

Background

Accumulating evidence indicates alterations in lipid metabolism and lipid composition in neoplastic tissue. Earlier nuclear magnetic resonance studies showed that the contents of major lipid groups, such as triacylglycerols, phospholipids and cholesterol, are changed in colon cancer tissue.

Methods

In this study, a more detailed analysis of lipids in cancer and tumor adjacent tissues from colorectal cancer patients, using liquid chromatography–mass spectrometry, allowed for comparison of 199 different lipids between cancer tissue and tumor adjacent tissue using principal component analysis.

Results

Significant differences were found in 67 lipid compounds between the two types of tissue; many of these lipid compounds are bioactive lipids such as ceramides, lysophospholipids or sterols and can influence the development of cancer. Additionally, increased levels of phospholipids and sphingolipids were present, which are major components of the cell membrane, and increases in these lipids can lead to changes in cell membrane properties.

Conclusions

This study showed that many complex lipids are significantly increased or decreased in colon cancer tissue, reflecting significant alterations in lipid metabolism. This knowledge can be used for the selection of potential molecular targets of novel anticancer strategies based on the modulation of lipid metabolism and the composition of the cell membrane in colorectal cancer cells.

Background

Colorectal cancer (CRC), according to the 2020 GLOBOCAN statistics, is among the top three most frequently diagnosed types of cancer and most fatal cancers in people of both sexes worldwide [1]. Despite the favorable effects of early screening and appropriate surveillance in developed nations, disparities due to socioeconomic factors and an alarming increase in CRC diagnosis in patients under 50 years continue to make CRC a considerable global public health issue [1, 2]. Progress in decreasing the burden of CRC through improved prevention or treatment needs detailed insight into metabolic alterations associated with this malignancy.

Because of advances in sample preparation methods and analytical techniques [3], lipid analysis has emerged as a useful tool in cancer research [4,5,6]. In particular, the rapid development of mass spectrometry methods enables increasingly sensitive and precise analysis [7]. Dysregulation of lipid metabolism has emerged as one of the most prominent phenotypic hallmarks of cancer [5]. Lipids are a complex group of biomolecules with varying structures and functions, and their role in cellular processes cannot be overstated. They play a role in energy metabolism and membrane formation, are precursors for the synthesis of signaling molecules and are even involved in the regulation of gene expression via epigenetic modulations [4, 5, 8]. Changes in lipid composition in biofluids and tissues are persistently associated with CRC [6], and lipid metabolism is being explored as a potential therapeutic target for CRC [4, 9] and in biomarker discovery [10,11,12,13].

Despite this knowledge, evidence on the exact nature of lipid alterations in colorectal cancer tissues is unclear. Intracellular accumulation of lipid droplets (LDs), organelles rich in neutral lipids, mainly triacylglycerols (TGs) and cholesteryl esters (CEs), has been reported in a number of neoplastic processes, and LDs play a role in a number of cancer metabolic hallmarks, such as hypoxia, death evasion and cell proliferation and inflammation [14]. The demonstration that LDs are a major site of cyclooxygenase-2 (COX-2) activity and prostaglandin E2 (PGE2) production in CRC cells highlighted the functional significance of LDs in cancer, and a higher number of LDs was detected in CRC tumor tissue than in adjacent normal tissue [15]. Interestingly, investigation of CRC cell lines revealed that although differentiated tumor cells contain greater amounts of LDs than normal epithelial cell lines, the greatest amount of LDs was found in CRC stem cell lines [16]. Furthermore, the tumorigenic potential of cancer stem cells was linked to overaccumulation of LDs, strengthening the idea that LDs are important in carcinogenesis. Wu et al. described the association between CRC progression and overaccumulation of LDs in tumor-associated macrophages, which increase the ability of tumors to grow and metastasize [17]. Accumulation of LDs in CRC cells was also found to promote CRC chemoresistance [18]. Taken together, these results point to LD metabolism as a potential therapeutic target. However, despite reports of LD accumulation in CRC tissue/cells, CRC tissue seems to be characterized by a paradoxically lower content of TGs, an LD-associated lipid group, than normal, cancer-adjacent tissue [19,20,21]. In addition, in a previous study, the total lipid content in CRC tissues was lower than that in tumor adjacent healthy mucosa [21]. These disparities highlight the gaps in understanding the role of LDs in cancer and emphasize the need for further investigation.

Phospholipid (PL) analysis is also increasingly performed in cancer lipidomic studies. PLs are the basic components of cellular membranes and thereby affect many membrane-associated processes, e.g., regulation of homeostasis, cell adhesion and migration; cellular signaling; cell-cell interactions; vesicular trafficking; and apoptosis [6, 22]. Alterations in the composition and distribution of PLs in cells, tissues and biofluids are associated with cancer [22] and have been explored as potential diagnostic or prognostic biomarkers in a variety of cancers, such as breast cancer [23], prostate cancer [24], lung cancer [25] and ovarian cancer [26]. PL analysis has also been applied in CRC to investigate the suitability of model colon cell lines [27] and three-dimensional culture systems [13] for lipidomic analysis in CRC studies. Accumulation of PL species reflects the increased amounts of polyunsaturated fatty acids (PUFAs), a phenomenon that has previously been observed in CRC tissues [28]. PLs, whose change in abundance is associated with CRC, include lysophospholipids (LPLs), an important group of signaling lipids [12, 29], and ether lipids, which can function as endogenous antioxidants [29]. Characterization of PL profiles in CRC tissues by imaging enabled researchers to localize some phosphatidylcholine species in colorectal cancer tissue regions and establish differences between tumor-adjacent and tumor-remote tissues [30] and was also applied for in vivo CRC phenotyping [20]. Interestingly, matrix assisted laser desorption and ionization (MALDI) mass spectrometry imaging also revealed distinct PL signatures that are able to discriminate between six different types of cancer microenvironments, therefore suggesting the possibility of a distinct lipogenic mechanism involved in these malignant processes [31].

Altered lipid metabolism in cancer cells is a potential molecular target of anticancer therapy. The best-known strategies are directed toward the fatty acid (FA) synthesis pathway. The well-known strategy of inhibition of fatty acid synthase (FASN) and the efficacy of the FASN inhibitor TVB-2640 have been tested in phase I and II clinical trials [32]. Additionally, orlistat, an anti-obesity drug, which also inhibits FASN activity, has shown anticancer effects in CRC cells [33]. However, other lipids may also be molecular targets for anticancer therapy. Related strategies include decreasing the cholesterol content [34], modulating lipid domains in cell membranes [35], and targeting membrane fluidity by modulating the lipid composition [36]. Thus, a thorough understanding of lipid dysregulation in CRC is crucial. To this end, lipid composition between tumor adjacent colon mucosa and cancer tissue in CRC patients was analyzed by liquid chromatography–mass spectrometry (LC-MS) and compared.

Methods

Patients

This LC-MS study was conducted on tissue samples obtained during surgical resection from patients with CRC who were included in previous investigations of FA profiles by gas chromatography–mass spectrometry (GC-MS) [28] and lipid groups by nuclear magnetic resonance (NMR) [21] to obtain more accurate data on abnormalities in complex lipids in tumor tissue. In the present study, 10 patients were included with T2-T4 CRC according to the TNM classification with a mean age of 68.4 ± 9.02 years and a mean body mass index (BMI) of 29.0 ± 4.27, characteristics of each patient are included in Supplementary Table 1. The included patients underwent primary resection of the large bowel without neoadjuvant chemo- or radiotherapy. The samples were collected from the tumor and tumor adjacent, micro and macroscopically normal large intestinal mucosa within the resection margin immediately after surgical resection. Each sample was divided into two parts. Recently, a representative photographs of H&E-stained tumor adjacent and cancer tissues from this group of patients were published [21]. The part of the sample designated for the lipidomic study was frozen in liquid nitrogen immediately after collection and stored at − 80 °C until analysis. The other part was examined histopathologically to confirm or exclude cancer tissue.

Lipids extraction

Samples were prepared as follows: aliquots of 50 mg of tumor and tumor adjacent tissue were homogenized in a chloroform-methanol mixture (1:1, v/v), saline was added, and the organic phase was collected and dried under a nitrogen stream. Prior to analysis, lipids were reconstituted in isopropanol to a final concentration of 1 mg/ml and passed through 0.2-μm PET filters.

Analysis by liquid chromatography–mass spectrometry (LC-MS)

The procedure for LC-MS analysis followed a method modified from Ulmer et al. [37]. The high-performance LC-MS system employed consisted of an HCT Ultra spectrometer (Bruker Daltonics, Billerica, Massachusetts, US) with an ESI source coupled with an Agilent 1200 liquid chromatograph (Agilent Technologies, Santa Clara, California, US). Chromatographic separation was conducted on a ReproSil-Pur Basic-C18 column (5 μm, 150 × 4.6 mm; Dr. Maisch GmbH, Ammerbuch, Germany). Phase A consisted of 10 mM ammonium formate in acetonitrile-water (60:40, v/v) with 0.1% formic acid, and phase B consisted of 10 mM ammonium formate in isopropanol-acetonitrile-water (90:8:2, v/v/v) with 0.1% formic acid. The employed gradient was as follows: 0 min – 45% B, 15 min – 75% B, 20 min – 95% B, 30 min – 95% B, 33 min – 45% B, and 38 min – 45% B. The flow was set at 0.4 ml/min, and the injection volume was 10 μl. Spectra were acquired in positive ESI mode, the capillary voltage was set at 136 V, the scanned mass range was 50–1500 m/z, and the accumulation time was 200,000 ms. Representative total ion chromatograms of tumor and tumor adjacent tissues are shown in Supplementary Fig. 1. The HPLC-MS method was tested using a mixture of standards consisting of TG (18:1/18:1/18:1), TG (16:0/16:0/16:0), cholesterol, ceramides (Cers) Cer(d18:1/6:0), Cer(d18:1/18:1(9Z)), sphingosine-1-phosphate, phosphatidylcholine (PC) (18:1/16:0) and palmitic acid (purchased from Sigma Aldrich, St. Louis, MO, USA) in switched polarity mode to determine retention time windows for lipid groups.

Data processing and statistical analysis

All spectra used for lipid identification were imported into SimLipid® 6.03 (PREMIER Biosoft, San Francisco, CA, USA) and preprocessed to exclude regions from 0 to 2.5 min and 28–38 min (column equilibration time). Lipid identification was done using high throughput MS lipid search, performed in the positive ion mode for [M + H], [M + NH4] ions, m/z tolerance 0.5 Da, in three time windows: 2–8 min, phosphatidylcholines (PC), phosphatidylethanolamines (PE), phosphatidylglycerols (PG), phosphatidylserine (PS); 8–23 min, glycerophospholipids, glycerolipids, sphingolipids, sterols; 22–28 min, glycerolipids, sterols, and results were reviewed manually. The m/z of observed adducts are presented in Supplementary Table 2. A peaklist alignment was performed, with maximum retention drift time across the runs 0.25 min, m/z tolerance of 0.05 and RT error tolerance set to the value of 0.2, peaks observed in < 50% of the samples were excluded. The data were normalized using the total response sum, log transformed and were subjected to Pareto scaling. Multivariate analysis and statistical data analysis were performed on a set of 199 unique assigned lipids in SIMCA software (version 16 Sartorius Stedim Data Analytics AB, Umeå, Sweden), only lipids that were present in at least 50% of the samples (both tumor and tumor adjacent tissues) were included in the analysis. The graphical representation of the results as a principal component analysis (PCA) biplot was constructed from the first two components, the software performs PCA model cross validation using the approach described by Eastment & Krzanowski [38]. Univariate analysis was performed with paired, two-tailed Student’s t-test.

Results

The method used allowed the detection of 199 different lipid species. PCA of the whole set of detected lipids showed that the lipid profile was different between tumor adjacent and CRC tissues (Fig. 1), although some overlapping was present.

Fig. 1
figure 1

PCA plot for all analyzed lipid species

Then, the sum of the signal intensities of the detected compounds within various groups of complex lipids between tumor adjacent mucosa and cancer tissue was compared. Among acylglycerols, the amounts of monoacylglycerols (MGs), diacylglycerols (DGs), and TGs were lower in tumor tissue than in tumor adjacent tissue (Fig. 2A-C). By contrast, the amounts of lipids forming cell membranes - PLs, LPLs, Cers and sphingolipids (SPLs) - were higher in tumor tissue (Fig. 2D-G). Only the amounts of sterols were similar between tumor tissue and tumor adjacent mucosa, with a slight increasing trend in tumor tissue (Fig. 2H).

Fig. 2
figure 2

The sum of the signal intensities of the detected compounds within various groups of complex lipids. Values are mean ± SEM. P-value from paired, two-tailed t-Student’s test

The results for individual species that were significantly different between tumor and tumor adjacent tissues are presented in Table 1, whereas the results for the whole set of detected lipids are presented in Supplementary Table 2. Only one MG (18:1) was detected that was significantly lower in tumor tissue. Among the 29 detected DGs, 11 were significantly different between tumor and tumor adjacent tissue: 8 were lower and 3 were higher in tumor tissue. Among the 37 detected TGs, 21 were significantly higher in tumor tissue, and none were significantly higher in tumor adjacent tissue. There was no significant correlation between the p values of the differences and the length or degree of saturation of the FAs forming acylglycerols. Among the 53 detected PLs, the signals of 17 were significantly higher and none were lower in tumor tissue than in tumor adjacent tissue. The phospholipids whose amounts were significantly higher in tumor tissue included 7 PCs, 3 PEs, 2 PSs and one PG. Additionally, ether phospholipids, namely, one 1-(1Z-alkenyl),2-acylglycerophosphate, one 1-alkyl,2-acylglycerophosphate and two 1-alkyl,2-acylglycerophosphocholines, were more abundant in tumor tissue. 30 LPLs were identified, and among them, 6 were significantly more abundant in tumor tissue: one lysophosphatidylcholine (LPC), 2 lysophosphatidylethanolamines (LPEs), 2 lysophosphatidylglycerols (LPGs) and 1 lysophosphatidylserine (LPS). Among identified 34 SPLs, 8 were significantly higher in tumor tissue than in tumor adjacent colon mucosa: 4 Cers, 2 sphingomyelins (SMs) and 2 sphingosines. Finally, 15 sterols were detected, including 8 CEs, but unfortunately, this method was not able to detect free cholesterol, which is a main component of the cell membrane. Among the detected sterols, the amounts of three were significantly different in tumor tissue compared to tumor adjacent tissue. The amounts of 22:3 cholesteryl ester and 3,5-cholestadien-7-one were higher, whereas the signal of 24,25-epoxy-cholesterol was lower, in tumor tissue (Table 1).

Table 1 Species of lipids significantly different between tumor and tumor adjacent tissues of CRC patients

Discussion

This study showed that the lipidome of CRC tissue is different from that of tumor adjacent mucosa; however, not all complex lipids exhibited significant differences between these two types of tissue in the studied group of patients. The PCA model revealed that tumor adjacent and tumor tissue cluster separately, although the separation between these two groups was not complete. This can possibly be attributed to heterogeneity of tissues of each patient, since two components of the PCA model account for 59.9% of total variance (Fig. 1). This tendency to separate based on lipid profiles contrasts the results of a recent study by Wang et al. [29] who did not observe separation of normal mucosa and tumor based on two component PCA (39.45% of total variance). Among the 199 identified lipids, the amounts of 67 were significantly different between tumor adjacent and cancer tissue. The results suggest that the direction of changes depends on the role played by individual groups of lipids in CRC cells. Acylglycerols constitute an energy depot in lipid droplets that is used for energy generation in these metabolically active cells, whereas PLs, SPLs and free cholesterol are cell membrane components that are urgently needed during CRC cell proliferation. Comparison of these respective groups of lipids showed that in cancer cells, the levels of energy-providing acylglycerols are lower, whereas the levels of membrane-building lipids are higher. These results are in agreement with a previous 1H-NMR study, which also showed decreased TG and increased PL, SM and free cholesterol contents in tumor tissue compared to tumor adjacent colon mucosa [21]. However, the present study provided much more detailed data on specific lipids; moreover, additional groups of lipids, including MG and DGs, LPLs and Cers, were detected.

Thus far, the reports on the role of abundance of neutral lipids in CRC seem inconclusive. The elevated content of neutral lipid bearing LDs was previously associated with CRC [15, 16, 39]. While some studies indicate that the TG levels in cancerous tissue were significantly lower than in paracancerous/tumor adjacent tissue of CRC patients [19, 21, 40], another found no significant differences in TG content and LDs abundance and distribution [29]. Moreover, the reports differ with regards to CRC advancement and TG levels, with one study reporting higher levels of TGs in early stage tumors [19], and another enrichment of TG in T3 tumors [40]. Hama et al. described significant decrease of TGs with long-chain FA moieties, which form a majority of TG fraction, while simultaneously reported the elevation of TGs containing very-long chain FAs [41]. Here, the sum levels of triglycerides were significantly lower in tumor tissues. MG and DG are both precursors of TG synthesis by acyltransferases and products of TG hydrolysis by lipases. All these acylglycerols are finally hydrolyzed to produce glycerol and FAs, which, after activation by acyl-CoA synthetase and transport to mitochondria with the participation of the carnitine palmitoyltransferase 1 (CPT1), can be used for energy production by beta-oxidation to acetyl-CoA, which is then oxidized to CO2 in the Krebs cycle. A previous study showed increased expression of CPT1 in cancer tissue [21], which suggests that oxidation of FAs stored as TG in lipid droplets is a possible reason for the decrease in TG. The simultaneous decrease in MG and DG shown in this study supports this hypothesis. The lack of correlation between the p values of the differences and the length or degree of saturation of FAs forming acylglycerols suggests that there is no preference regarding the length or saturation of FAs during TG hydrolysis. However, the increased levels of PUFAs and saturated FAs (SFAs) and decreased levels of monounsaturated FAs (MUFAs) in CRC cells [28] suggest that SFAs and PUFAs are directed toward cell membrane synthesis, whereas MUFAs are preferentially used as an energy source and undergo beta-oxidation. It is also worth mentioning that MUFAs, which can be synthesized by stearoyl-CoA desaturase-1 (SCD1) in human cells [42], are the main component of TG and that SCD1 is overexpressed in many types of cancer, including CRC [6, 43,44,45]. It seems that overexpression of both SCD1 and FASN [33] is associated with increased production of TG, which include MUFAs that are conversely largely oxidized to provide energy to cancer cells. Another source of FAs for beta-oxidation may be the import of free FAs that are released from adipocytes adjacent to cancer cells [46]. DG is also a signaling molecule associated with the development of various cancers, but its role is associated with the activity of diacylglycerol kinases that target DG originating from hydrolysis of cell membrane PLs by phospholipase C [47], whereas the pool of DG in lipid droplets seems to be associated with the synthesis/degradation of TG in cancer cells. Recently, the issue of concurrent fatty acid synthesis and oxidation was extensively discussed in a review published by De Olivera et al. [48]. This group proposed the existence of two types of mitochondria—lipogenic mitochondria, which release citrate into the cytosol to fuel lipogenesis, and fatty acid oxidizing mitochondria, which produce ATP from fat [48].

In addition, PLs, LPLs and SPLs are cell membrane components, and their levels were increased in tumor tissue. This phenomenon is probably associated with increased synthesis of plasma membranes in rapidly proliferating cancer cells, as was suggested in a previous study [21]. Guo et al. [31], using MALDI-MSI, studied various cancers, including samples from six patients with CRC. Their results differed from those of described in this study; they found increases in MUFAs and decreases in PUFAs, as well as increases in PC containing MUFAs, but decreases in PC, phosphatidylinositol (PI) and PE containing PUFAs in cancer tissue. Surprisingly, they suggested that the synthesis of PUFAs from 18:1 FAs is downregulated [31], whereas this process is not possible in human tissues due to the lack of delta-12 and delta-15 desaturases [49]. By contrast, in this study, all significantly different PLs, both those containing MUFAs and those containing PUFAs, were more abundant in cancer tissue. This phenomenon may be associated with increased lipogenesis, lipolysis and exogenous FA intake—processes that provide FAs for the synthesis of membrane phospholipids and other membrane lipids [50]. The last process is especially important in the case of exogenous PUFAs (18:2 n-6 and 18:3 n-3) [28], which cannot be synthesized in human cells [49]. Increased levels of ether phospholipids were also detected. Ether lipids are increased in cancers and correlate with greater aggressiveness [51]. However, their exact role in promoting cancer progression is not known. It has been shown that they regulate ion channels, which may constitute the mechanism underlying the regulation of cell proliferation [52]. Likewise, the exact nature of sphingolipid metabolism in carcinogenic processes is not yet clear, but seems to be dependent on acyl-chain composition [53]. In current study, the difference in total Cer amounts between tumor and tumor adjacent tissue (Fig. 1) are in agreement with previous reports of elevated Cer synthases expression levels [54], as well as total Cer content in CRC tissues [29, 53].

The overall LPL content, in which LPCs are major contributors, detected here was increased, which contrasts with the results of Wang et al. [29], wherein they reported significant decrease across LPL classes (LPC, LPE) and increase in lysophosphatidylinositol (LPI). Contrary to that, Kitamura et al. [55] reported significantly higher levels of LPI and LPS in colon cancer tissue, and higher, although not statistically significant, levels of LPC, LPE and LPG. The results of present study support Kitamura et al. findings, the direction of change of particular species: LPE(16:1), LPE(18:2), LPG(20:4) (Table 1), aligns with their results, albeit the upregulation of these species in Kitamura et al. study lacked statistical significance, which may possibly arise due to small number of patients in both studies. The upregulation of LPLs seems surprising, considering that overexpression of enzymes LPCAT1 and LPCAT2, which re-acylate LPCs into PCs, was observed in CRC cells [18, 56]. However, these enzymes are associated with LDs, which exhibit high inter-individual variation that may possibly explain these opposing findings [29]. LPLs are precursors of lysophosphatidic acid (LPA), which is a signaling molecule and can inhibit p53 activity by activating the LPA receptor [57]. In addition, some ceramides activate p53 pathways [57]. Thus, both LPLs and ceramides may influence cancer cell proliferation and apoptosis; however, since both groups of lipids are increased in cancer tissue, it is hard to speculate about the combined effect of these compounds on p53 pathways. Additionally, LPE can act as a signaling molecule, stimulating the migration and invasion of human ovarian cancer cells by interacting with G protein-coupled receptors [58]. In addition, LPS is associated with inflammation [59], which is a characteristic factor for the development of CRC [6]. Moreover, their proinflammatory properties increase with the FA chain length [60], and in current study a significantly increased amount of LPS containing 20:0 FAs was present. SPLs are involved in the regulation of cell differentiation, proliferation and apoptosis, as well as drug resistance [61,62,63]; thus, increased levels of lipids from this group may also be important for CRC cell metabolism.

Among sterols, only 3 metabolites were significantly different between tumor and cancer tissues. One of them was 22:3 CE, which was increased in tumor tissue, but all seven other CEs did not differ significantly between tumor adjacent and tumor tissue, and the total sterol amount was not significantly different. The largely preserved CE profile is consistent with findings of Hama et al. [41], although it must be noted that in this study number of detected CE species was limited. CEs are strongly hydrophobic molecules and are located in lipid droplets. Unfortunately, the procedure used did not allow for detection of free cholesterol, which is an important component of cell membranes, by this method, but a previous study using 1H-NMR, showed that the level of free cholesterol is significantly higher in CRC tissue [21], which is consistent with the concept of the increased content of membrane lipids in CRC cells. Increased free cholesterol levels in cancer cell membranes are associated with increased levels of lipid rafts and increased resistance to anticancer drugs [46]. Another significant metabolite may contribute to increased levels of free cholesterol. 24(S),25-epoxycholesterol is a negative regulator of HMG-CoA reductase (HMGCR), the rate-limiting enzyme in cholesterol synthesis, that acts by binding to Liver X Receptor (LXR) [64]. A previous study showed increased expression of HMGCR in CRC tissue [21]. The present analysis revealed a significantly (approximately 40%) lower level of 24(S),25-epoxycholesterol in tumor tissue than in tumor adjacent tissue; thus, this decreased level may be one of the reasons for the elevated cholesterol synthesis and elevated free cholesterol levels in CRC cells. Interestingly, upregulation of 24(R/S),25-epoxycholesterol inhibits the proliferation of gastric cancer cells [65]. Another interesting sterol is 3,5 cholestadiene-7-one, which is a product of membrane cholesterol autooxidation [66], a process caused by oxidative stress that is present in cancer tissue [48].

Study strengths and limitations

The most important advantage of this study was further discovering and confirming the altered lipid metabolism in CRC tissue. The results partly confirmed our previous studies [11, 21, 28, 67], but also provided a lot of new data. The advantage of this study was also using the matched tumor adjacent, micro and macroscopically normal colon mucosa and tumor samples from the same patients. Although the issue of field cancerization has been raised before [68] the paired tissue samples seem to be the most suitable controls, given the apparent heterogeneity of tissue samples [29, 53].

The limitations of the study include small number of samples, which did not allow to analyze correlations between demographic and clinical data and the results of lipidomic analysis. Also, the limited sensitivity of the LC-MS setup necessarily restrained lipid identification reporting up to lipid species/bond type level, due to the lack of MS/MS data. On the other hand, using this relatively simple approach, the trend for separation between tumor and tumor adjacent tissue could be observed and the ease of the procedure could be advantageous when expanding the cohort size.

Conclusions

In conclusion, this study identified many complex lipids that are significantly increased or decreased in CRC tissue. These data extend the knowledge on alterations in the composition of CRC tissue. This knowledge can be used for the selection of potential molecular targets of novel anticancer strategies based on the modulation of lipid metabolism and the composition of the cell membrane in CRC cells. A detailed understanding of the observed alterations requires analyses of tissues from primary tumors stratified by the clinical stage of CRC. It seems probable that the lipid composition changes with the accumulation of mutations and progression from adenoma to carcinoma (in a well-described adenoma-to-carcinoma sequence) and further through the clinical stages of CRC. Defining alterations in locally advanced (stage I and II), regionally advanced (stage III) and disseminated (stage IV) disease would enable an understanding of the role of lipids in CRC progression. In future, this knowledge could also transfer into better CRC classification and possibly better inclusion criteria for adjuvant therapy if significant association between lipid profiles and CRC prognosis could be established. Moreover, it would be of interest to define alterations in individual cancer grades (from grade 1, where cancer cells look similar to tumor adjacent mucosal cells (well differentiated) to grade 3, where cancer cells look very abnormal (poorly differentiated).

Availability of data and materials

The dataset analyzed in this study can be reasonably obtained from the corresponding author.

Abbreviations

BMI:

body mass index

CE:

cholesteryl ester

Cer:

ceramide

COX:

cyclooxygenase

CPT1:

carnitine palmitoyltransferase 1

CRC:

colorectal cancer

DG:

diacylglycerol

FA:

fatty acid

FASN:

fatty acid synthase

GC-MS:

gas chromatography–mass spectrometry

HMGCR:

HMG-CoA reductase

LC-MS:

liquid chromatography–mass spectrometry

LD:

lipid droplet

LPC:

lysophosphatidylcholine

LPE:

lysophosphatidylethanolamine

LPG:

lysophosphatidylglycerol

LPI:

lysophosphatidylinositol

LPL:

lysophospholipid

LPS:

lysophosphatidylserine

LXR:

Liver X Receptor

MG:

monoacylglycerol

MALDI:

matrix assisted laser desorption and ionization

MUFA:

monounsaturated fatty acid

NMR:

nuclear magnetic resonance

PC:

phosphatidylcholine

PCA:

principal component analysis

PE:

phosphatidylethanolamine

PG:

phosphatidylglycerol

PGE2:

prostaglandin E2

PI:

phosphatidylinositol

PL:

phospholipid

PS:

phosphatidylserine

PUFA:

polyunsaturated fatty acid

SCD1:

stearoyl-CoA desaturase-1

SFA:

saturated fatty acid

SM:

sphingomyelin

SPL:

sphingolipid

TAG:

triacylglycerol

References

  1. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;0:caac.21660.

    Google Scholar 

  2. Siegel RL, Miller KD, Goding Sauer A, Fedewa SA, Butterly LF, Anderson JC, et al. Colorectal cancer statistics, 2020. CA Cancer J Clin. 2020;70(3):145–64. https://doi.org/10.3322/caac.21601.

    Article  PubMed  Google Scholar 

  3. Liakh I, Sledzinski T, Kaska L, Mozolewska P, Mika A. Sample preparation methods for Lipidomics approaches used in studies of obesity. Molecules. 2020;25(22):5307. https://doi.org/10.3390/molecules25225307.

    Article  CAS  PubMed Central  Google Scholar 

  4. Zaytseva Y. Lipid metabolism as a targetable metabolic vulnerability in colorectal Cancer. Cancers. 2021;13(2):301. https://doi.org/10.3390/cancers13020301.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Snaebjornsson MT, Janaki-Raman S, Schulze A. Greasing the Wheels of the Cancer Machine: The Role of Lipid Metabolism in Cancer. Cell Metab. Elsevier Inc. 2020;31:62–76.

    Article  CAS  Google Scholar 

  6. Pakiet A, Kobiela J, Stepnowski P, Sledzinski T, Mika A. Changes in lipids composition and metabolism in colorectal cancer: a review. Lipids Health Dis. 2019;18(1):29. https://doi.org/10.1186/s12944-019-0977-8.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Mika A, Sledzinski T, Stepnowski P. Current Progress of lipid analysis in metabolic diseases by mass spectrometry methods. Curr Med Chem. 2019;26(1):60–103. https://doi.org/10.2174/0929867324666171003121127.

    Article  CAS  PubMed  Google Scholar 

  8. Sabari BR, Zhang D, Allis CD, Zhao Y. Metabolic regulation of gene expression through histone acylations. Nat Rev Mol Cell Biol. 2017;18(2):90–101. https://doi.org/10.1038/nrm.2016.140.

    Article  CAS  PubMed  Google Scholar 

  9. Mozolewska P, Duzowska K, Pakiet A, Mika A, Śledziński T. Inhibitors of fatty acid synthesis and oxidation as potential anticancer agents in colorectal Cancer treatment. Anticancer Res. 2020;40(9):4843–56. https://doi.org/10.21873/anticanres.14487.

    Article  CAS  PubMed  Google Scholar 

  10. Zhang L, Chen B, Zhang J, Li J, Yang Q, Zhong Q, et al. Serum polyunsaturated fatty acid metabolites as useful tool for screening potential biomarker of colorectal cancer. Prostaglandins Leukot Essent Fat Acids. 2017;120:25–31. https://doi.org/10.1016/j.plefa.2017.04.003.

    Article  CAS  Google Scholar 

  11. Mika A, Kobiela J, Czumaj A, Chmielewski M, Stepnowski P, Sledzinski T. Hyper-elongation in colorectal Cancer tissue – cerotic acid is a potential novel serum metabolic marker of colorectal malignancies. Cell Physiol Biochem. 2017;41(2):722–30. https://doi.org/10.1159/000458431.

    Article  CAS  PubMed  Google Scholar 

  12. Li F, Qin X, Chen H, Qiu L, Guo Y, Liu H, et al. Lipid profiling for early diagnosis and progression of colorectal cancer using direct-infusion electrospray ionization Fourier transform ion cyclotron resonance mass spectrometry. Rapid Commun Mass Spectrom. 2013;27(1):24–34. https://doi.org/10.1002/rcm.6420.

    Article  CAS  PubMed  Google Scholar 

  13. Hiraide T, Ikegami K, Sakaguchi T, Morita Y, Hayasaka T, Masaki N, et al. Accumulation of arachidonic acid-containing phosphatidylinositol at the outer edge of colorectal cancer. Sci Rep Nat Publ Group. 2016;6:29935.

    CAS  Google Scholar 

  14. Cruz ALS, Barreto D de A, NPB F, JPB V, Bozza PT. Lipid droplets: platforms with multiple functions in cancer hallmarks. Cell Death Dis. 2020;11:105.

    Article  Google Scholar 

  15. Accioly MT, Pacheco P, Maya-Monteiro CM, Carrossini N, Robbs BK, Oliveira SS, et al. Lipid bodies are reservoirs of Cyclooxygenase-2 and sites of prostaglandin-E 2 synthesis in Colon Cancer cells. Cancer Res. 2008;68(6):1732–40. https://doi.org/10.1158/0008-5472.CAN-07-1999.

    Article  CAS  PubMed  Google Scholar 

  16. Tirinato L, Liberale C, Di Franco S, Candeloro P, Benfante A, La Rocca R, et al. Lipid droplets: a new player in colorectal Cancer stem cells unveiled by spectroscopic imaging. Stem Cells. 2015;33(1):35–44. https://doi.org/10.1002/stem.1837.

    Article  CAS  PubMed  Google Scholar 

  17. Wu H, Han Y, Rodriguez Sillke Y, Deng H, Siddiqui S, Treese C, et al. Lipid droplet-dependent fatty acid metabolism controls the immune suppressive phenotype of tumor-associated macrophages. EMBO Mol Med. 2019;11:1–17.

    Article  CAS  Google Scholar 

  18. Cotte AK, Aires V, Fredon M, Limagne E, Derangère V, Thibaudin M, et al. Lysophosphatidylcholine acyltransferase 2-mediated lipid droplet production supports colorectal cancer chemoresistance. Nat Commun. 2018;9(1):322. https://doi.org/10.1038/s41467-017-02732-5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Zhang X. Lipid levels in serum and cancerous tissues of colorectal cancer patients. World J Gastroenterol. 2014;20(26):8646–52. https://doi.org/10.3748/wjg.v20.i26.8646.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Alexander J, Louise Gildea B, Julia Balog B, Abigail Speller B, James McKenzie B, Laura Muirhead B, et al. A novel methodology for in vivo endoscopic phenotyping of colorectal cancer based on real-time analysis of the mucosal lipidome: a prospective observational study of the iKnife. Surg Endosc. 2017;31(3):1361–70. https://doi.org/10.1007/s00464-016-5121-5.

    Article  PubMed  Google Scholar 

  21. Mika A, Pakiet A, Czumaj A, Kaczynski Z, Liakh I, Kobiela J, et al. Decreased triacylglycerol content and elevated contents of cell membrane lipids in colorectal Cancer tissue: a Lipidomic study. J Clin Med. 2020;9(4):1095. https://doi.org/10.3390/jcm9041095.

    Article  CAS  PubMed Central  Google Scholar 

  22. Bandu R, Mok HJ, Kim KP. Phospholipids as cancer biomarkers: mass spectrometry-based analysis. Mass Spectrom Rev. 2018;37(2):107–38. https://doi.org/10.1002/mas.21510.

    Article  CAS  PubMed  Google Scholar 

  23. Budczies J, Denkert C, Müller BM, Brockmöller SF, Klauschen F, Györffy B, et al. Remodeling of central metabolism in invasive breast cancer compared to normal breast tissue – a GC-TOFMS based metabolomics study. BMC Genomics. 2012;13(1):334. https://doi.org/10.1186/1471-2164-13-334.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Goto T, Terada N, Inoue T, Kobayashi T, Nakayama K, Okada Y, et al. Decreased expression of lysophosphatidylcholine (16:0/OH) in high resolution imaging mass spectrometry independently predicts biochemical recurrence after surgical treatment for prostate cancer. Prostate. 2015;75(16):1821–30. https://doi.org/10.1002/pros.23088.

    Article  CAS  PubMed  Google Scholar 

  25. Lee GK, Lee HS, Park YS, Lee JH, Lee SC, Lee JH, et al. Lipid MALDI profile classifies non-small cell lung cancers according to the histologic type. Lung Cancer. 2012;76(2):197–203. https://doi.org/10.1016/j.lungcan.2011.10.016.

    Article  PubMed  Google Scholar 

  26. Meleh M, Požlep B, Mlakar A, Meden-Vrtovec H, Zupančič-Kralj L. Determination of serum lysophosphatidic acid as a potential biomarker for ovarian cancer. J Chromatogr B. 2007;858(1-2):287–91. https://doi.org/10.1016/j.jchromb.2007.08.008.

    Article  CAS  Google Scholar 

  27. Hofmanová J, Slavík J, Ovesná P, Tylichová Z, Dušek L, Straková N, et al. Phospholipid profiling enables to discriminate tumor- and non-tumor-derived human colon epithelial cells: Phospholipidome similarities and differences in colon cancer cell lines and in patient-derived cell samples. PLoS One. 2020;15(1):e0228010. https://doi.org/10.1371/journal.pone.0228010.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Mika A, Kobiela J, Pakiet A, Czumaj A, Sokołowska E, Makarewicz W, et al. Preferential uptake of polyunsaturated fatty acids by colorectal cancer cells. Sci Rep. 2020;10(1):1954. https://doi.org/10.1038/s41598-020-58895-7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Wang Y, Hinz S, Uckermann O, Hönscheid P, von Schönfels W, Burmeister G, et al. Shotgun lipidomics-based characterization of the landscape of lipid metabolism in colorectal cancer. Biochim Biophys Acta - Mol Cell Biol Lipids. 1865;2020:158579.

    Google Scholar 

  30. Mirnezami R, Spagou K, Vorkas PA, Lewis MR, Kinross J, Want E, et al. Chemical mapping of the colorectal cancer microenvironment via MALDI imaging mass spectrometry (MALDI-MSI) reveals novel cancer-associated field effects. Mol Oncol. 2014;8(1):39–49. https://doi.org/10.1016/j.molonc.2013.08.010.

    Article  CAS  PubMed  Google Scholar 

  31. Guo S, Wang Y, Zhou D, Li Z. Significantly increased monounsaturated lipids relative to polyunsaturated lipids in six types of cancer microenvironment are observed by mass spectrometry imaging. Sci Rep. 2014;4:5959.

    Article  CAS  Google Scholar 

  32. Schcolnik-Cabrera A, Chávez-Blanco A, Domínguez-Gómez G, Taja-Chayeb L, Morales-Barcenas R, Trejo-Becerril C, et al. Orlistat as a FASN inhibitor and multitargeted agent for cancer therapy. Expert Opin Investig Drugs. 2018;27(5):475–89. https://doi.org/10.1080/13543784.2018.1471132.

    Article  CAS  PubMed  Google Scholar 

  33. Czumaj A, Zabielska J, Pakiet A, Mika A, Rostkowska O, Makarewicz W, et al. In vivo effectiveness of orlistat in the suppression of human colorectal cancer cell proliferation. Anticancer Res. 2019;39(7):3815–22. https://doi.org/10.21873/anticanres.13531.

    Article  CAS  PubMed  Google Scholar 

  34. Srivatsav AT, Mishra M, Kapoor S. Small-molecule modulation of lipid-dependent cellular processes against Cancer: fats on the gunpoint. Biomed Res. 2018:6437371.

  35. Escribá PV. Membrane-lipid therapy: a new approach in molecular medicine. Trends Mol. Med. 2006;12:34–43.

    Google Scholar 

  36. Zalba S, ten Hagen TLM. Cell membrane modulation as adjuvant in cancer therapy. Cancer Treat Rev. 2017;52:48–57. https://doi.org/10.1016/j.ctrv.2016.10.008.

    Article  CAS  PubMed  Google Scholar 

  37. Ulmer CZ, Patterson RE, Koelmel JP, Garrett TJ, Yost RA. A robust Lipidomics workflow for mammalian cells, plasma, and tissue using liquid-chromatography high-resolution tandem mass spectrometry. Physiol Behav. 2017;176:91–106.

    Google Scholar 

  38. Eastment HT, Krzanowski WJ. Cross-validatory choice of the number of components from a principal component analysis. Technometrics. 1982;24(1):73–7. https://doi.org/10.1080/00401706.1982.10487712.

    Article  Google Scholar 

  39. Zaytseva YY, Harris JW, Mitov MI, Kim JT, Butterfield DA, Lee EY, et al. Increased expression of fatty acid synthase provides a survival advantage to colorectal cancer cells via upregulation of cellular respiration. Oncotarget. 2015;6(22):18891–904. https://doi.org/10.18632/oncotarget.3783.

    Article  PubMed  PubMed Central  Google Scholar 

  40. Mirnezami R, Jiménez B, Li JV, Kinross JM, Veselkov K, Goldin RD, et al. Rapid diagnosis and staging of colorectal Cancer via high-resolution magic angle spinning nuclear magnetic resonance (HR-MAS NMR) spectroscopy of intact tissue biopsies. Ann Surg. 2014;259(6):1138–49. https://doi.org/10.1097/SLA.0b013e31829d5c45.

    Article  PubMed  Google Scholar 

  41. Hama K, Fujiwara Y, Hayama T, Ozawa T, Nozawa K, Matsuda K, et al. Very long-chain fatty acids are accumulated in triacylglycerol and nonesterified forms in colorectal cancer tissues. Sci Rep. 2021;11(1):6163. https://doi.org/10.1038/s41598-021-85603-w.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Mika A, Kaska L, Korczynska J, Mirowska A, Stepnowski P, Proczko M, et al. Visceral and subcutaneous adipose tissue stearoyl-CoA desaturase-1 mRNA levels and fatty acid desaturation index positively correlate with BMI in morbidly obese women. Eur J Lipid Sci Technol. 2015;117(7):926–32. https://doi.org/10.1002/ejlt.201400372.

    Article  CAS  Google Scholar 

  43. Presler M, Wojtczyk-Miaskowska A, Schlichtholz B, Kaluzny A, Matuszewski M, Mika A, et al. Increased expression of the gene encoding stearoyl-CoA desaturase 1 in human bladder cancer. Mol Cell Biochem. 2018;447(1-2):217–24. https://doi.org/10.1007/s11010-018-3306-z.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Swierczynski J, Hebanowska A, Sledzinski T. Role of abnormal lipid metabolism in development, progression, diagnosis and therapy of pancreatic cancer. World J Gastroenterol. 2014;20(9):2279–303. https://doi.org/10.3748/wjg.v20.i9.2279.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Mika A, Sledzinski T. Alterations of specific lipid groups in serum of obese humans: a review. Obes Rev. 2017;18(2):247–72. https://doi.org/10.1111/obr.12475.

    Article  CAS  PubMed  Google Scholar 

  46. Beloribi-Djefaflia S, Vasseur S, Guillaumond F. Lipid metabolic reprogramming in cancer cells. Oncogenesis. 2016;5(1):e189. https://doi.org/10.1038/oncsis.2015.49.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Mérida I, Torres-Ayuso P, Ávila-Flores A, Arranz-Nicolás J, Andrada E, Tello-Lafoz M, et al. Diacylglycerol kinases in cancer. Adv Biol Regul. 2017;63:22–31. https://doi.org/10.1016/j.jbior.2016.09.005.

    Article  CAS  PubMed  Google Scholar 

  48. De Oliveira MP, Liesa M. The role of mitochondrial fat oxidation in Cancer cell proliferation and survival. Cells. 2020;9(12):2600. https://doi.org/10.3390/cells9122600.

    Article  CAS  PubMed Central  Google Scholar 

  49. Czumaj A, Śledziński T. Biological role of unsaturated fatty acid desaturases in health and disease. Nutrients. 2020;12(2):356. https://doi.org/10.3390/nu12020356.

    Article  CAS  PubMed Central  Google Scholar 

  50. Baenke F, Peck B, Miess H, Schulze A. Hooked on fat: the role of lipid synthesis in cancer metabolism and tumour development. Dis Model Mech. 2013;6(6):1353–63. https://doi.org/10.1242/dmm.011338.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Dean JM, Lodhi IJ. Structural and functional roles of ether lipids. Protein Cell. 2018;9(2):196–206. https://doi.org/10.1007/s13238-017-0423-5.

    Article  CAS  PubMed  Google Scholar 

  52. Fontaine D, Figiel S, Félix R, Kouba S, Fromont G, Mahéo K, et al. Roles of endogenous ether lipids and associated PUFAs in the regulation of ion channels and their relevance for disease. J Lipid Res. 2020;61(6):840–58. https://doi.org/10.1194/jlr.RA120000634.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Markowski AR, Błachnio-Zabielska AU, Guzińska-Ustymowicz K, Markowska A, Pogodzińska K, Roszczyc K, et al. Ceramides profile identifies patients with more advanced stages of colorectal Cancer. Biomolecules. 2020;10(4):632. https://doi.org/10.3390/biom10040632.

    Article  CAS  PubMed Central  Google Scholar 

  54. Jang S, Park W, Min H, Kwon T, Baek S, Hwang I, et al. Altered mRNA expression levels of the major components of sphingolipid metabolism, ceramide synthases and their clinical implication in colorectal cancer. Oncol Rep. 2018;40(6):3489–500. https://doi.org/10.3892/or.2018.6712.

    Article  CAS  PubMed  Google Scholar 

  55. Kitamura C, Sonoda H, Nozawa H, Kano K, Emoto S, Murono K, et al. The component changes of lysophospholipid mediators in colorectal cancer. Tumor Biol. 2019;41(5):101042831984861. https://doi.org/10.1177/1010428319848616.

    Article  CAS  Google Scholar 

  56. Mansilla F, da Costa K-A, Wang S, Kruhøffer M, Lewin TM, Ørntoft TF, et al. Lysophosphatidylcholine acyltransferase 1 (LPCAT1) overexpression in human colorectal cancer. J Mol Med. 2009;87(1):85–97. https://doi.org/10.1007/s00109-008-0409-0.

    Article  CAS  PubMed  Google Scholar 

  57. Lee SC, Lin K-H, Balogh A, Norman DD, Bavaria M, Kuo B, et al. Dysregulation of lysophospholipid signaling by p53 in malignant cells and the tumor microenvironment. Cell Signal. 2021;78:109850. https://doi.org/10.1016/j.cellsig.2020.109850.

    Article  CAS  PubMed  Google Scholar 

  58. Park KS, Lee HY, Lee SY, Kim M-K, Kim SD, Kim JM, et al. Lysophosphatidylethanolamine stimulates chemotactic migration and cellular invasion in SK-OV3 human ovarian cancer cells: involvement of pertussis toxin-sensitive G-protein coupled receptor. FEBS Lett. 2007;581(23):4411–6. https://doi.org/10.1016/j.febslet.2007.08.014.

    Article  CAS  PubMed  Google Scholar 

  59. Frasch SC, Bratton DL. Emerging roles for lysophosphatidylserine in resolution of inflammation. Prog Lipid Res. 2012;51(3):199–207. https://doi.org/10.1016/j.plipres.2012.03.001.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Khandelwal N, Shaikh M, Mhetre A, Singh S, Sajeevan T, Joshi A, et al. Fatty acid chain length drives lysophosphatidylserine-dependent immunological outputs. Cell Chem Biol. 2021;28:1–11.

    Article  Google Scholar 

  61. Adada M, Luberto C, Canals D. Inhibitors of the sphingomyelin cycle: Sphingomyelin synthases and sphingomyelinases. Chem Phys Lipids. 2016;197:45–59. https://doi.org/10.1016/j.chemphyslip.2015.07.008.

    Article  CAS  PubMed  Google Scholar 

  62. Lewis AC, Wallington-Beddoe CT, Powell JA, Pitson SM. Targeting sphingolipid metabolism as an approach for combination therapies in haematological malignancies. Cell Death Discov. 2018;4(1):72. https://doi.org/10.1038/s41420-018-0075-0.

    Article  PubMed  PubMed Central  Google Scholar 

  63. Kartal Yandım M, Apohan E, Baran Y. Therapeutic potential of targeting ceramide/glucosylceramide pathway in cancer. Cancer Chemother Pharmacol. 2013;71(1):13–20. https://doi.org/10.1007/s00280-012-1984-x.

    Article  CAS  PubMed  Google Scholar 

  64. Wong J, Quinn CM, Brown AJ. Synthesis of the oxysterol, 24(S), 25-epoxycholesterol, parallels cholesterol production and may protect against cellular accumulation of newly-synthesized cholesterol. Lipids Health Dis. 2007;6(1):10. https://doi.org/10.1186/1476-511X-6-10.

    Article  PubMed  PubMed Central  Google Scholar 

  65. Guo F, Hong W, Yang M, Xu D, Bai Q, Li X, et al. Upregulation of 24(R/S),25-epoxycholesterol and 27-hydroxycholesterol suppresses the proliferation and migration of gastric cancer cells. Biochem Biophys Res Commun. 2018;504(4):892–8. https://doi.org/10.1016/j.bbrc.2018.09.058.

    Article  CAS  PubMed  Google Scholar 

  66. Sevanian A, McLeod LL. Cholesterol autoxidation in phospholipid membrane bilayers. Lipids Springer-Verlag. 1987;22:627–36.

    CAS  Google Scholar 

  67. Mika A, Duzowska K, Halinski LP, Pakiet A, Czumaj A, Rostkowska O, et al. Rearrangements of blood and tissue fatty acid profile in colorectal Cancer - molecular mechanism and diagnostic potential. Front Oncol. 2021;11:689701. https://doi.org/10.3389/fonc.2021.689701.

    Article  PubMed  PubMed Central  Google Scholar 

  68. Aran D, Camarda R, Odegaard J, Paik H, Oskotsky B, Krings G, et al. Comprehensive analysis of normal adjacent to tumor transcriptomes. Nat Commun. 2017;8:1–14.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

The study was funded by National Science Centre of Poland, grant number 2016/22/E/NZ4/00665 and Medical University of Gdansk, grants number ST-40 and ST-89.

Author information

Authors and Affiliations

Authors

Contributions

AM and TS designed the study. Research material was collected by JK and OR and analysis was performed by AP, KS and AM. TS, AM and AP analyzed and interpreted the data. TS and AP wrote the manuscript. JK, AM and TS reviewed the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Tomasz Sledzinski.

Ethics declarations

Ethics approval and consent to participate

The protocol of the study was compliant with the Declaration of Helsinki of the World Medical Association and was granted approval from the Local Bioethics Committee at the Medical University of Gdansk (decision no. NKBN/487/2015). Written informed consent was obtained from all the patients prior to the study.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Additional file 1: Figure S1.

Total ion chromatograms of representative samples from tumor adjacent (A) and tumor (B) tissues. Abbreviations: CE, cholesteryl ester; Cer, ceramide; DG, diacylglycerol; LPL, lysophospholipids; MG, monoacylglycerol; PA, phosphatidic acid; PC, phosphatidylcholine; PE, phosphatidylethanolamine; PG, phosphatidylglycerol; PHL, phospholipid; PS, phosphatidylserine; SM, sphingomyelin; TG, triacylglycerol.

Additional file 2: Table S1.

Characteristics of colorectal cancer patients included in the study. Abbreviations: BPH, benign prostatic hyperplasia; COPD; chronic obstructive pulmonary disease; HT, hypertension; RA, rheumatoid arthritis; T2DM, type 2 diabetes.

Additional file 3: Table S2.

Species of lipids detected in tumor and tumor adjacent tissues of CRC patients. Values are mean signal intensity (million units) ± SEM. P-value from paired, two-tailed t-Student’s test.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Pakiet, A., Sikora, K., Kobiela, J. et al. Alterations in complex lipids in tumor tissue of patients with colorectal cancer. Lipids Health Dis 20, 85 (2021). https://doi.org/10.1186/s12944-021-01512-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12944-021-01512-x

Keywords