Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Role of sphingosine kinase and sphingosine-1-phosphate receptor in the liver pathology of mice infected with Plasmodium berghei ANKA

  • Tachpon Techarang,

    Roles Conceptualization, Data curation, Formal analysis, Investigation, Methodology, Validation, Writing – original draft

    Affiliation Department of Medical Science, School of Medicine, Walailak University, Nakhon Si Thammarat, Thailand

  • Pitchanee Jariyapong,

    Roles Conceptualization, Data curation, Formal analysis, Investigation, Methodology, Validation, Writing – original draft, Writing – review & editing

    Affiliation Department of Medical Science, School of Medicine, Walailak University, Nakhon Si Thammarat, Thailand

  • Chuchard Punsawad

    Roles Conceptualization, Data curation, Formal analysis, Funding acquisition, Investigation, Methodology, Project administration, Supervision, Validation, Writing – original draft, Writing – review & editing

    chuchard.pu@wu.ac.th

    Affiliations Department of Medical Science, School of Medicine, Walailak University, Nakhon Si Thammarat, Thailand, Research Center in Tropical Pathobiology, Walailak University, Nakhon Si Thammarat, Thailand

Abstract

Decreased serum sphingosine 1-phosphate (S1P) has been reported in severe malaria patients, but the expression of receptors and enzymes associated with S1P has not been investigated in the liver of malaria patients. Therefore, this study aimed to investigate the expression of sphingosine kinase (SphK) and S1P receptors (S1PRs) in the liver of malaria-infected mice. C57BL/6 male mice were divided into a control group (n = 10) and a Plasmodium berghei (PbA)-infected group (n = 10). Mice in the malaria group were intraperitoneally injected with 1×106 P. berghei ANKA-infected red blood cells, whereas control mice were intraperitoneally injected with normal saline. Liver tissues were collected on Day 13 of the experiment to evaluate histopathological changes by hematoxylin and eosin staining and to investigate SphK and S1PR expression by immunohistochemistry and real-time PCR. Histological examination of liver tissues from the PbA-infected group revealed sinusoidal dilatation, hemozoin deposition, portal tract inflammation and apoptotic hepatocytes, which were absent in the control group. Immunohistochemical staining showed significant increases in the expression of SphK1 and SphK2 and significant decreases in the expression of S1PR1, S1PR2, and S1PR3 in the endothelium, hepatocytes, and Kupffer cells in liver tissue from the PbA-infected group compared with the control group. Real-time PCR analysis showed the upregulation of SphK1 and the downregulation of S1PR1, S1PR2, and S1PR3 in the liver in the PbA-infected group compared with the control group. In conclusion, this study demonstrates for the first time that SphK1 mRNA expression is upregulated and that S1PR1, S1PR2, and S1PR3 expression is decreased in the liver tissue of PbA-infected mice. Our findings suggest that the decreased levels of S1PR1, S1PR2, and S1PR3 might play an important role in liver injury during malaria infection.

Introduction

Malaria is a life-threatening infectious disease caused by Plasmodium parasites that remains a cause of morbidity and mortality in humans; there were 229 million clinical cases in 2019 and 409,000 deaths worldwide, of which 67% were children under 5 years of age [1]. Plasmodium parasites are typically transmitted to humans by the bite of an infected female Anopheles mosquito. Plasmodium parasites travel to the liver, where they mature and reproduce [2]. There are several severe complications of malaria, such as cerebral malaria, acute lung injury/acute respiratory distress syndrome, acute kidney injury, hypoglycemia, lactic acidosis, severe anemia and acute liver failure [3]. The liver is the first important organ affected during the hepatic stage of infection and is the site in which malaria sporozoites develop into merozoites. Protozoan parasites damage hepatocytes in all stages of the malaria parasite life cycle, especially in the hepatic stage [4]. In addition, liver damage may be caused by the binding of infected red blood cells to endothelial cells, leading to the obstruction of hepatic sinusoids and intrahepatic blood flow. It has been reported that histopathological changes in liver tissue during malaria infection are characterized by Kupffer cell hyperplasia, abundant hemozoin pigment in hyperplastic Kupffer cells and hepatocytes, inflammatory cell infiltration in the portal tract, (especially neutrophils and monocytes), fatty liver changes, liver fibrosis, cholestasis, bile stasis, granulomatous lesions and malarial nodules [46].

Sphingosine 1-phosphate (S1P) is a lysophospholipid mediator that can stimulate a wide variety of intracellular and extracellular responses, such as cell proliferation, differentiation, migration, contraction, and survival, as well as the immune response [710]. S1P is released after the phosphorylation of sphingosine by sphingosine kinase (SphK) [11]. Indeed, previous studies revealed that S1P signaling is mediated by a family of five high-affinity G protein-coupled receptors, including S1PR1, S1PR2, S1PR3, S1PR4, and S1PR5 [1113]. However, only three receptors for S1P are expressed in the liver: S1PR1, S1PR2, and S1PR3. SphKs and S1PRs have been shown to have novel therapeutic potential in the treatment of cancer, gastrointestinal disease, obesity, and diabetes mellitus and its associated complications [9, 10, 14, 15]. In addition, the overexpression of SphK1 in macrophages conferred resistance to mycobacterial infection [16]. In the context of malaria, a recent investigation showed reduced serum levels of S1P in patients with severe malaria [17, 18]. Therefore, we hypothesized that SphK/S1PR plays a role in the severity of malaria and liver damage. However, there have been no reports on the expression of receptors and enzymes associated with S1P in the liver in the context of malaria.

Therefore, this study aimed to investigate histopathological changes and the expression of SphKs (SphK1 and SphK2) and S1P receptors (S1PR1, S1PR2, and S1PR3) in the liver tissue of malaria-infected mice compared with control mice. Understanding the pathophysiological role of the receptors and enzymes associated with S1P may provide new insights into malaria pathogenesis and new therapeutic options for the treatment of malaria infection.

Methods

Ethics statement

This study protocol was reviewed and approved by the Animal Ethics Committee of Walailak University (Clearance no. 007/2018). All protocols in this study were conducted in accordance with the relevant guidelines and regulations for using animals in compliance with Animal Research: Reporting of In Vivo Experiments (ARRIVE) guidelines. Animals were anesthetized with pentobarbital to minimize pain and suffering.

Animal preparation and experimental protocols

Male C57BL/6 mice (6–10 weeks old) were purchased from Nomura Siam International Co., Ltd. (Bangkok, Thailand). The mice were housed at 22–24°C in standard polycarbonate cages (5 animals per cage) on wood-shaving bedding with free access to food and fresh water and a standard 12 h light/dark cycle. Plasmodium berghei ANKA was obtained from BEI Resources (NIAID, NIH: P. berghei, Strain ANKA, MRA-311) and was contributed by Thomas F. McCutchan. Donor mice were inoculated with P. berghei ANKA-infected red blood cells, and blood was collected via cardiac puncture into a heparinized vacutainer tube for the induction of malaria in experimental mice [19]. For the malaria-infected group, the mice were intraperitoneally infected with 1×106 Plasmodium berghei ANKA (PbA)-infected red blood cells as previously described [20, 21]. Control mice were intraperitoneally administered 200 μl of saline. After infection, parasitemia levels were monitored daily using Giemsa-stained peripheral blood smears. On the 13th day after infection (DAI), the mice were sacrificed under anesthesia with an intraperitoneal injection of pentobarbital (60 mg/kg body weight) to minimize suffering. After opening the abdominal cavity, liver tissues were collected immediately for histopathological, immunohistochemical and real-time PCR analyses. Finally, the mice were intraperitoneally injected with an overdose of pentobarbital (150 mg/kg) for euthanasia.

Histopathological examination

Liver tissues were fixed in 10% formalin for 24–48 h, embedded in paraffin, sectioned (5 μm thickness) and stained with hematoxylin and eosin as previously described [22]. For histological assessments, alterations in four histological parameters (sinusoidal dilatation, hemozoin deposition, portal tract inflammation and apoptotic hepatocytes) were assessed under a light microscope at high magnification (400×) by two expert pathologists. Based on a previous study with modifications [4], the severity of each parameter was graded using a semiquantitative scoring system, as shown in Table 1. The total histological score was calculated as the sum of the scores for each of the four histological parameters and ranged from 0 to 12 points.

thumbnail
Table 1. Grading system for four main histological features to assess the severity of liver injury.

https://doi.org/10.1371/journal.pone.0266055.t001

Immunohistochemistry

Formalin-fixed, paraffin-embedded (FFPE) sections of liver tissue were deparaffinized in xylene and rehydrated in a series of ethanol solutions. FFPE tissue sections were incubated in 3% hydrogen peroxide to quench endogenous peroxidase activity. To improve antigen retrieval, the sections were incubated in Vector Antigen Unmasking Solution (Vector Laboratories Inc., USA) according to the manufacturer’s instructions. After being washed in Tris-buffered saline (TBS), the sections were blocked for 30 min in normal goat serum in TBS. FFPE tissue sections were further incubated for 1 h at room temperature with rabbit polyclonal antibodies against SphK1 or SphK2 (1:200; Ab71700 and Ab264042, Abcam, UK) or S1PR1, S1PR2 or S1PR3 (1:200; sc-25489, Santa Cruz Biotechnology, Inc., USA), followed by a 30 min incubation with diluted biotinylated secondary antibodies and a 30 min incubation with VECTASTAIN ABC Reagent (Vector Laboratories Inc., USA). Then, all FFPE tissue sections were incubated in DAB reagent (brown chromogen staining; Vector Laboratories Inc., USA), counterstained with hematoxylin, rehydrated, and mounted for light microscopy analysis. Negative controls were prepared in the same manner but without primary antibodies. To evaluate the expression of SphKs (SphK1 and SphK2) and S1P receptors (S1PR1, S1PR2 and S1PR3), the number of positive cells and the intensity of staining in 10 randomly selected microscopic fields of each immunostained section were determined at high magnification (400×). The expression of each protein was separately examined in endothelial cells, hepatocytes, and Kupffer cells. The percentage of positively stained cells for each protein marker was calculated by dividing the number of positive cells by the total cell count and multiplying this number by 100. The staining intensity was subjectively scored as follows: 0 = no staining, 1 = weakly positive, 2 = moderately positive, and 3 = strongly positive. Finally, the total score (TS) was calculated by multiplying the percentage of positive cells (%) and staining intensity (I), according to a previous study [23].

Real-time polymerase chain reaction (real-time PCR)

SphK and S1PR mRNA levels in liver tissue in the control and PbA-infected groups were measured by quantitative real-time PCR. RNA was extracted using TriPure isolation reagent (Roche, Mannheim, Germany) according to the manufacturer’s instructions. First strand cDNA was generated using an iScript™ cDNA Synthesis Kit (Bio–Rad, Philadelphia, PA). The reaction was incubated at 25°C for 5 min, 42°C for 30 min, and 85°C for 5 min. cDNA (50 ng) from each sample was subsequently amplified using gene-specific primer sets (Table 2). The thermal conditions consisted of initial denaturation at 94°C for 10 s, followed by 40 cycles of denaturation at 94°C for 30 s, annealing at 55°C for 30 s, and elongation at 72°C for 30 s, and a final elongation step at 72°C for 5 min. The relative expression of target genes was determined using the 2−ΔΔCt method and beta-actin as a comparator.

Statistical analysis

Statistical analysis was performed using IBM SPSS version 23.0 (SPSS, IL, USA). The normality of distribution was tested with the Kolmogorov–Smirnov test. Differences between groups (malaria-infected and control groups) were analyzed by a nonparametric Mann–Whitney U test. A p value < 0.05 was considered to indicate statistical significance.

Results

Histopathological examination

Histopathological analysis of liver tissues from the PbA-infected group (Fig 1A and 1C) showed sinusoidal dilatation, hemozoin deposition, portal tract inflammation and hepatocyte apoptosis compared to those in the control group (Fig 1B and 1C). Semiquantitative analysis demonstrated that the mean scores of all histological parameters were significantly higher in the PbA-infected group than in the control group (all p < 0.05) (Table 3). The histopathological change with the highest mean was hemozoin deposition (2.64 ± 0.13), followed by portal tract inflammation (1.40 ± 0.09).

thumbnail
Fig 1.

Histopathological changes in liver tissues from control (A) and PbA-infected mice (B). The histomorphological changes in PbA-infected mice include hemozoin deposition (indicated by yellow arrowheads) and apoptotic hepatocytes (indicated by black asterisks) around the central vein. Central vein (CV); Portal tract (PT). All images are shown at 200× magnification. Scale bar = 100 μm.

https://doi.org/10.1371/journal.pone.0266055.g001

thumbnail
Table 3. Mean scores of four main histological features in tissues from the control (n = 10) and PbA-infected groups (n = 10).

https://doi.org/10.1371/journal.pone.0266055.t003

SphK and S1PR expression as indicated by immunohistochemical staining

Immunohistochemical analysis of SphK and S1PR expression in liver tissues from control and PbA-infected mice is shown in Fig 2. Positive staining is indicated by brown. The number of positively stained cells and the staining intensity for each protein were independently examined in each individual cell type, including endothelial, epithelial, and Kupffer cells. The mean TS for SphK1 and SphK2 expression was significantly increased in endothelial cells, hepatocytes, and Kupffer cells in liver tissue from PbA-infected mice compared with control mice (p < 0.05). However, the mean TS for S1PR1, S1PR2, and S1PR3 was significantly decreased in endothelial cells, hepatocytes, and Kupffer cells in liver tissue from PbA-infected mice compared with control mice (p < 0.05) (Table 4).

thumbnail
Fig 2.

Immunoperoxidase staining for SphK and S1PR in liver tissues from control (A, C, E, G, I) and PbA-infected mice (B, D, F, H, J). Hepatocytes (H), Kupffer cells (K), sinusoids (S), and central veins (CVs). All images are shown at 200× magnification. Scale bar = 50 μm.

https://doi.org/10.1371/journal.pone.0266055.g002

thumbnail
Table 4. Mean TS for S1PR and SphK expression in liver tissues from the control (n = 10) and PbA-infected (n = 10) groups.

https://doi.org/10.1371/journal.pone.0266055.t004

SphK and S1PR mRNA levels as determined by real-time PCR

The expression of genes encoding enzymes and receptors associated with S31P in liver tissues from control and PbA-infected mice was investigated by real-time PCR (Fig 3). The expression of enzymes responsible for regulating the concentration of S1P, including SphK1 and SphK2, was measured in liver tissue from PbA-infected mice. Our results showed that SphK1 mRNA levels were upregulated approximately 0.5-fold in liver tissue from PbA-infected mice, whereas SphK2 mRNA levels did not change (Fig 3A). In addition, S1PR1 and S1PR2 mRNA levels were decreased in liver tissue from PbA-infected mice compared to control mice, whereas S1PR3 mRNA levels were significantly decreased compared to those in the control group (Fig 3B).

thumbnail
Fig 3. Relative SphK and S1PR mRNA expression as measured by real-time PCR.

Data are presented as the mean ± SD of triplicate experiments. *p < 0.05 compared with the control group.

https://doi.org/10.1371/journal.pone.0266055.g003

Discussion

The liver is one of the organs in which infected red blood cells preferentially accumulate to avoid immune system surveillance inside the spleen, and infected red blood cell sequestration within the liver endothelium is now emerging as another factor that promotes liver damage [2628]. In the present study, histopathological examination demonstrated that liver tissues from PbA-infected mice exhibited sinusoidal dilatation, hemozoin deposition, portal tract inflammation and hepatocyte apoptosis. These findings are consistent with previous studies showing evidence of portal tract inflammation and hemozoin pigment deposition in liver tissue from patients with severe P. falciparum malaria and hyperbilirubinemia [4] and from C57BL/6 mice infected with PbA [29], indicating that this murine model could be a potential resource for investigating the pathological mechanisms of liver damage caused by malaria infection.

S1P is a bioactive sphingolipid mediator that functions intracellularly as a second messenger and extracellularly as a ligand for specific G protein-coupled receptors [1113], affecting processes such as cell proliferation, differentiation, migration, contraction, and survival, as well as inflammation [710]. In the context of malaria, protozoan parasite infection can induce the release of proinflammatory cytokines and chemokines, resulting in parasite destruction and an inflammatory response [30, 31]. Our previous study reported that serum S1P concentrations were decreased in malaria patients infected with P. vivax and P. falciparum and that low levels of S1P were associated with the severity of malaria [18]. However, there have been no reports on the expression of SphK or S1PR in the liver during malaria infection. Herein, we investigated the expression of SphKs (SphK1 and 2) and S1PRs (S1PR1, 2, and 3) in liver tissue from mice infected with PbA. The present study demonstrated SphK and S1PR expression based on positive immunohistochemical staining in three liver cell types, including endothelial cells, hepatocytes, and Kupffer cells. Compared with control mice, PbA-infected mice showed a significant increase in the number of cells that were positively stained for both SphK1 and SphK2 and a significant decrease in the expression of all S1PRs in all cell types in liver tissue.

Normally, S1P is located in both intra- and extracellular regions [32]. This protein is exported from cells via transporters such as spinster homolog 2 (Spns2) on the endothelial surface to maintain circulating S1P concentrations [3234]. S1P is released into the extracellular space after the phosphorylation of sphingosine by SphKs. Therefore, the mRNA expression of receptors and enzymes responsible for S1P signaling was also investigated. The present study revealed that the mRNA expression of S1PRs, especially S1PR3, was downregulated, whereas only SphK1 mRNA was significantly upregulated in PbA-infected mice compared with control mice. These findings suggest the following. 1) When infected red blood cells rupture during parasite growth and differentiation [2], an important source of circulating S1P is needed due to a lack of cellular organelles [32, 35, 36], and S1P is released into the circulatory system. This protein can bind to plasma components such as high-density lipoprotein (HDL), low-density lipoprotein (LDL), and albumin [37] and thereby decrease S1P activity to stimulate S1PRs on the cell surface, leading to decreases in S1PR activation and expression. 2) Circulating S1P can induce apoptosis through G protein-coupled receptors on the cell surface via caspase-3 [38]. This signaling stimulates the production of inflammatory mediators, the expression of ICAM-1 and VCAM-1 on the cell surface, and the release of Weibel–Palade bodies, which are organelles in endothelial cells that store various proteins involved in inflammation, resulting in an inflammatory response and cell apoptosis [3840]. 3) The inflammatory response to protozoan parasite infection and hemozoin, a waste product of hemoglobin digestion by the Plasmodium parasite, induces leucocyte accumulation. This accumulation leads to the secretion of proinflammatory cytokines, such as tumor necrosis factor alpha (TNF-α), transforming growth factor beta (TGF-β), interferon gamma (IFN-γ), interleukin-1 (IL-1), and IL-6 [4143]. Some cytokines stimulate SphK1 activation, followed by the activation of RAS [44] and ERK1/2 and the transcription of NF-κB [45], resulting in inflammation. The inflammatory response to malaria destroys the Plasmodium parasite, but negative effects also occur, including cell membrane damage, especially in endothelial cells. Therefore, the expression of S1PRs on the cell surface was dramatically decreased in response to cell injury or cell death. This result suggested that S1PRs can be downregulated by cell injury, which might be stimulated by proinflammatory cytokines.

In addition, the current study demonstrated an increase in SphK1 in malaria-infected mice, which was consistent with previous reports on inflammatory conditions and cancer [9, 14, 15, 4648]. This finding may stem from inflammation-induced apoptosis. The potential role of SphK1 expression in this model might be the consequence of malaria infection. After malaria infection, the parasites travel to the liver, where they replicate to generate many schizonts in hepatocytes. This situation induces hepatocyte rupture, followed by merozoite release into circulation [2]. In addition, these merozoites can infect red blood cells, where they replicate and develop into the sexual form [2]. As a result, immunological responses are triggered, including the release of proinflammatory cytokines and white blood cell accumulation [30, 49, 50]. TNF-α and other proinflammatory cytokines (IL-1 and IL-6) have been shown to activate SphK1, leading to increased S1P synthesis and release [14]. Cytoplasmic S1P can be irreversibly degraded into ethanolamine phosphate and hexadecenal by the intracellular protein sphingosine phosphate lyase [51]. The hexadecenal molecule can form adducts with proteins, lipids, and DNA and inhibit histone deacetylases (HDACs) in the nucleus, thereby further promoting inflammatory responses in the cytoplasm [14]. In response to inflammation, proinflammatory cytokines stimulate sphingomyelin hydrolysis by alkaline sphingomyelinase [14]. This action initiates ceramide production from hepatocyte membranes. The increase in ceramide in hepatocytes leads to oxidative stress, proinflammatory cytokine secretion, and hepatocyte apoptosis [52]. Therefore, these proinflammatory cytokines can stimulate hepatocyte apoptosis, resulting in a decrease in S1PRs during SphK1 expression in malaria (Fig 4). In addition, SphK2 evokes apoptotic signaling by promoting the local production of S1P [10]. Alternately, the binding of released S1P and S1PRs initiates various intracellular downstream signaling pathways, such as the NF-κB, ERK1/2, and Rac/PLC pathways, which can induce cell survival, proliferation, and migration [10, 14]. A previous study reported that apoptosis stimulated SphK1 expression in Jurkat T cells and U937 monocytes [53]. In a final attempt to prevent cell death, a cell might increase the levels of SphK1 and S1P, which are survival mediators. In addition, the activation of intracellular SphK1 has been reported to protect pulmonary vascular endothelial cells during LPS-mediated inflammation in vivo [54]. This result suggested that SphK1 might play an important role in preventing cell injury under inflammatory conditions. Further studies should focus on the mechanism of SphK and S1PR signaling during liver injury caused by malaria infection.

thumbnail
Fig 4. A schematic illustration of SphK/S1PR signaling during malaria infection.

Plasmodium parasite-infected red blood cells induce the accumulation of macrophages, neutrophils, and monocytes, which further initiates the release of proinflammatory cytokines such as TNF-α, IL-1, and IL-6. These proinflammatory cytokines, especially TNF-α, bind to specific receptors on the cell membrane, which can activate sphingomyelin hydrolysis. Next, ceramide can be hydrolyzed to sphingosine by neutral ceramidase. Then, SphKs phosphorylate sphingosine to generate S1P, which directly induces NF-κB activation and translocates to the extracellular space, where it can bind S1PRs and stimulate intracellular downstream signaling, resulting in inflammation and cell survival, proliferation, and migration. In addition, S1P can be degraded into ethanolamine phosphate and hexadecenal; in particular, hexadecenal can stimulate further inflammation in the cytoplasm.

https://doi.org/10.1371/journal.pone.0266055.g004

Some limitations of this study should be noted. The duration of SphK and S1PR changes during malaria infection could not be determined. Therefore, further kinetic studies on SphK and S1PR expression are required to characterize the time-dependent expression of these proteins. Second, the protein levels of SphKs in this animal model were not assessed. Thus, these protein levels should be monitored in future experiments.

Conclusion

The present study indicated that liver tissue from malaria-infected mice has decreased expression of S1PR1, S1PR2, and S1PR3 and increased expression of SphK1. These findings suggest that alterations in S1PR1, S1PR2, S1PR3 and SphK1 are involved in the inflammatory response and apoptosis and play important roles in the pathogenesis of malaria in the liver.

Acknowledgments

The authors would like to acknowledge the laboratory workers at the Animal Experiment Building, Walailak University, Thailand, for facilitating all procedures performed in mice. We also thank the staff members at the Department of Tropical Pathology, Faculty of Tropical Medicine, Mahidol University, Thailand, for helping with histological processing.

References

  1. 1. WHO. World Malaria Report 2020. 2020.
  2. 2. CDC. Malaria 2019 [cited 2020 20 May]. Available from: https://www.cdc.gov/malaria/index.html.
  3. 3. White NJ. The Treatment of Malaria. N Engl J Med. 1996;335(11):800–6. pmid:8703186
  4. 4. Viriyavejakul P, Khachonsaksumet V, Punsawad C. Liver changes in severe Plasmodium falciparum malaria: histopathology, apoptosis and nuclear factor kappa B expression. Malar J. 2014;13:106–. pmid:24636003
  5. 5. WHO. Severe falciparum malaria. Trans R Soc Trop Med Hyg. 2000;94:1–90. pmid:10748883
  6. 6. Baheti R, Laddha P, Gehlot RS. Liver Involvement in Falciparum Malaria–A Histopathological Analysis. J Indian Acad Clin Med. 2003;4(1):34–8.
  7. 7. Hannun YA, Obeid LM. Principles of bioactive lipid signalling: lessons from sphingolipids. Nature Reviews Molecular Cell Biology. 2008;9(2):139–50. pmid:18216770
  8. 8. Hla T. Physiological and pathological actions of sphingosine 1-phosphate. Semin Cell Dev Biol. 2004;15(5):513–20. pmid:15271296
  9. 9. Sukocheva OA. Expansion of Sphingosine Kinase and Sphingosine-1-Phosphate Receptor Function in Normal and Cancer Cells: From Membrane Restructuring to Mediation of Estrogen Signaling and Stem Cell Programming. Int J Mol Sci. 2018;19(2). pmid:29385066
  10. 10. Ng ML, Wadham C, Sukocheva OA. The role of sphingolipid signalling in diabetes‑associated pathologies (Review). Int J Mol Med. 2017;39(2):243–52. pmid:28075451
  11. 11. Spiegel S, Milstien S. Sphingosine-1-phosphate: an enigmatic signalling lipid. Nature Reviews Molecular Cell Biology. 2003;4:397. pmid:12728273
  12. 12. Gardell SE, Dubin AE, Chun J. Emerging medicinal roles for lysophospholipid signaling. Trends Mol Med. 2006;12(2):65–75. pmid:16406843
  13. 13. Ishii I, Fukushima N, Ye X, Chun J. Lysophospholipid Receptors: Signaling and Biology. Annu Rev Biochem. 2004;73(1):321–54. pmid:15189145
  14. 14. Sukocheva OA, Furuya H, Ng ML, Friedemann M, Menschikowski M, Tarasov VV, et al. Sphingosine kinase and sphingosine-1-phosphate receptor signaling pathway in inflammatory gastrointestinal disease and cancers: A novel therapeutic target. Pharmacol Ther. 2020;207:107464. pmid:31863815
  15. 15. Sukocheva O, Wadham C, Xia P. Estrogen defines the dynamics and destination of transactivated EGF receptor in breast cancer cells: Role of S1P3 receptor and Cdc42. Exp Cell Res. 2013;319:455–65. pmid:23142484
  16. 16. Prakash H, Lüth A, Grinkina N, Holzer D, Wadgaonkar R, Gonzalez AP, et al. Sphingosine Kinase-1 (SphK-1) Regulates Mycobacterium smegmatis Infection in Macrophages. PLoS One. 2010;5(5):e10657. pmid:20498849
  17. 17. Finney CAM, Hawkes CA, Kain DC, Dhabangi A, Musoke C, Cserti-Gazdewich C, et al. S1P Is Associated with Protection in Human and Experimental Cerebral Malaria. Mol Med. 2011;17(7–8):717–25. pmid:21556483
  18. 18. Punsawad C, Viriyavejakul P. Reduction in serum sphingosine 1-phosphate concentration in malaria. PLoS One. 2017;12(6):e0180631. pmid:28666023
  19. 19. Kaewdana K, Chaniad P, Jariyapong P, Phuwajaroanpong A, Punsawad C. Antioxidant and antimalarial properties of Sophora exigua Craib. root extract in Plasmodium berghei-infected mice. Trop Med Health. 2021;49(1):24. pmid:33741053
  20. 20. Epiphanio S, Campos MG, Pamplona A, Carapau D, Pena AC, Ataide R, et al. VEGF promotes malaria-associated acute lung injury in mice. PLoS Pathog. 2010;6(5):e1000916. pmid:20502682
  21. 21. El-Assaad F, Wheway J, Mitchell AJ, Lou J, Hunt NH, Combes V, et al. Cytoadherence of Plasmodium berghei-infected red blood cells to murine brain and lung microvascular endothelial cells in vitro. Infect Immun. 2013;81(11):3984–91. pmid:23940206
  22. 22. Punsawad C, Viriyavejakul P, Setthapramote C, Palipoch S. Enhanced expression of Fas and FasL modulates apoptosis in the lungs of severe P. falciparum malaria patients with pulmonary edema. Int J Clin Exp Pathol. 2015;8(9):10002–13. pmid:26617708
  23. 23. Punsawad C, Maneerat Y, Chaisri U, Nantavisai K, Viriyavejakul P. Nuclear factor kappa B modulates apoptosis in the brain endothelial cells and intravascular leukocytes of fatal cerebral malaria. Malar J. 2013;12:260. pmid:23890318
  24. 24. Michaud J, Kohno M, Proia RL, Hla T. Normal acute and chronic inflammatory responses in sphingosine kinase 1 knockout mice. FEBS Letters. 2006;580(19):4607–12. pmid:16876794
  25. 25. Li C, Jiang X, Yang L, Liu X, Yue S, Li L. Involvement of Sphingosine 1-Phosphate (SIP)/S1P(3) Signaling in Cholestasis-Induced Liver Fibrosis. The American Journal of Pathology. 2009;175(4):1464–72. pmid:19729475
  26. 26. Medeiros MM, da Silva HB, Reis AS, Barboza R, Thompson J, Lima MR, et al. Liver accumulation of Plasmodium chabaudi-infected red blood cells and modulation of regulatory T cell and dendritic cell responses. PLoS One. 2013;8(11):e81409. pmid:24312297
  27. 27. Fonager J, Pasini EM, Braks JA, Klop O, Ramesar J, Remarque EJ, et al. Reduced CD36-dependent tissue sequestration of Plasmodium-infected erythrocytes is detrimental to malaria parasite growth in vivo. J Exp Med. 2012;209(1):93–107. pmid:22184632
  28. 28. Franke-Fayard B, Fonager J, Braks J, Khan S, Janse J, Chris. Sequestration and Tissue Accumulation of Human Malaria Parasites: Can We Learn Anything from Rodent Models of Malaria? PLoS Pathog. 2010;6(9):e1001032. pmid:20941396
  29. 29. Kim J, Wang S, Lee C, Sung S, Shin Y, Song KS, et al. Blood-Stage Plasmodium Berghei ANKA Infection Promotes Hepatic Fibrosis by Enhancing Hedgehog Signaling in Mice. Cell Physiol Biochem. 2018;50(4):1414–28. pmid:30355912
  30. 30. Mbengue B, Niang B, Niang MS, Varela ML, Fall B, Fall MM, et al. Inflammatory cytokine and humoral responses to Plasmodium falciparum glycosylphosphatidylinositols correlates with malaria immunity and pathogenesis. Immun Inflamm Dis. 2015;4(1):24–34. pmid:27042299
  31. 31. Griffith JW, Sun T, McIntosh MT, Bucala R. Pure Hemozoin is inflammatory in vivo and activates the NALP3 inflammasome via release of uric acid. J Immunol. 2009;183(8):5208–20. pmid:19783673
  32. 32. Obinata H, Hla T. Sphingosine 1-phosphate and inflammation. Int Immunol. 2019;31(9):617–25. pmid:31049553
  33. 33. Venkataraman K, Lee Y-M, Michaud J, Thangada S, Ai Y, Bonkovsky HL, et al. Vascular endothelium as a contributor of plasma sphingosine 1-phosphate. Circ Res. 2008;102(6):669–76. pmid:18258856
  34. 34. Kawahara A, Nishi T, Hisano Y, Fukui H, Yamaguchi A, Mochizuki N. The sphingolipid transporter spns2 functions in migration of zebrafish myocardial precursors. Science. 2009;323(5913):524–7. pmid:19074308
  35. 35. Hänel P, Andréani P, Gräler MH. Erythrocytes store and release sphingosine 1-phosphate in blood. FASEB J. 2007;21(4):1202–9. pmid:17215483
  36. 36. Bode C, Sensken SC, Peest U, Beutel G, Thol F, Levkau B, et al. Erythrocytes serve as a reservoir for cellular and extracellular sphingosine 1-phosphate. J Cell Biochem. 2010;109(6):1232–43. pmid:20186882
  37. 37. Murata N, Sato K, Kon J, Tomura H, Yanagita M, Kuwabara A, et al. Interaction of sphingosine 1-phosphate with plasma components, including lipoproteins, regulates the lipid receptor-mediated actions. Biochem J. 2000;352 Pt 3(Pt 3):809–15. pmid:11104690
  38. 38. Davaille J, Li L, Mallat A, Lotersztajn S. Sphingosine 1-Phosphate Triggers Both Apoptotic and Survival Signals for Human Hepatic Myofibroblasts*. J Biol Chem. 2002;277(40):37323–30. pmid:12138095
  39. 39. Yogi A, Callera Glaucia E, Aranha Anna B, Antunes Tayze T, Graham D, McBride M, et al. Sphingosine-1-Phosphate-Induced Inflammation Involves Receptor Tyrosine Kinase Transactivation in Vascular Cells. Hypertension. 2011;57(4):809–18. pmid:21383307
  40. 40. van Hooren KW, Spijkers LJ, van Breevoort D, Fernandez-Borja M, Bierings R, van Buul JD, et al. Sphingosine-1-phosphate receptor 3 mediates sphingosine-1-phosphate induced release of weibel-palade bodies from endothelial cells. PLoS One. 2014;9(3):e91346. pmid:24632890
  41. 41. Deroost K, Tyberghein A, Lays N, Noppen S, Schwarzer E, Vanstreels E, et al. Hemozoin induces lung inflammation and correlates with malaria-associated acute respiratory distress syndrome. Am J Respir Cell Mol Biol. 2013;48(5):589–600. pmid:23328641
  42. 42. Maknitikul S, Luplertlop N, Grau GER, Ampawong S. Dysregulation of pulmonary endothelial protein C receptor and thrombomodulin in severe falciparum malaria-associated ARDS relevant to hemozoin. PLoS One. 2017;12(7):e0181674–e. pmid:28732053
  43. 43. Maknitikul S, Luplertlop N, Chaisri U, Maneerat Y, Ampawong S. Featured Article: Immunomodulatory effect of hemozoin on pneumocyte apoptosis via CARD9 pathway, a possibly retarding pulmonary resolution. Exp Biol Med. 2018;243(5):395–407. pmid:29402133
  44. 44. Shu X, Wu W, Mosteller RD, Broek D. Sphingosine Kinase Mediates Vascular Endothelial Growth Factor-Induced Activation of Ras and Mitogen-Activated Protein Kinases. Mol Cell Biol. 2002;22(22):7758. pmid:12391145
  45. 45. Pitson SM, Moretti PAB, Zebol JR, Xia P, Gamble JR, Vadas MA, et al. Expression of a Catalytically Inactive Sphingosine Kinase Mutant Blocks Agonist-induced Sphingosine Kinase Activation: A DOMINANT-NEGATIVE SPHINGOSINE KINASE*. J Biol Chem. 2000;275(43):33945–50. pmid:10944534
  46. 46. Li J, Zhang B, Bai Y, Liu Y, Zhang B, Jin J. Upregulation of sphingosine kinase 1 is associated with recurrence and poor prognosis in papillary thyroid carcinoma. Oncol Lett. 2019;18(5):5374–82. pmid:31620198
  47. 47. Xia P, Wang L, Moretti PA, Albanese N, Chai F, Pitson SM, et al. Sphingosine kinase interacts with TRAF2 and dissects tumor necrosis factor-alpha signaling. J Biol Chem. 2002;277(10):7996–8003. pmid:11777919
  48. 48. Viriyavejakul P, Punsawad C. Overexpression of Sphingosine Kinase-1 and Sphingosine-1-Phosphate Receptor-3 in Severe Plasmodium falciparum Malaria with Pulmonary Edema. BioMed Research International. 2020;2020:3932569. pmid:32185202
  49. 49. Clark I, Cowden WB, Butcher GA, Hunt NH. Possible roles of tumor necrosis factor in the pathology of malaria. The American Journal of Pathology. 1987;129(1):192–9. pmid:3661678
  50. 50. Alves MFC, Santos-Neto LL, Junqueira MIM, Costa CE. Cytokines and dysregulation of the immune response in human malaria. Mem Inst Oswaldo Cruz. 1992;87:331–6. pmid:1343709
  51. 51. Serra M, Saba JD. Sphingosine 1-phosphate lyase, a key regulator of sphingosine 1-phosphate signaling and function. Adv Enzyme Regul. 2010;50(1):349–62. pmid:19914275
  52. 52. Hajduch E, Lachkar F, Ferré P, Foufelle F. Roles of Ceramides in Non-Alcoholic Fatty Liver Disease. Journal of Clinical Medicine. 2021;10(4):792. pmid:33669443
  53. 53. Gude DR, Alvarez SE, Paugh SW, Mitra P, Yu J, Griffiths R, et al. Apoptosis induces expression of sphingosine kinase 1 to release sphingosine-1-phosphate as a "come-and-get-me" signal. FASEB J. 2008;22(8):2629–38. pmid:18362204
  54. 54. Wadgaonkar R, Patel V, Grinkina N, Romano C, Liu J, Zhao Y, et al. Differential regulation of sphingosine kinases 1 and 2 in lung injury. American Journal of Physiology-Lung Cellular and Molecular Physiology. 2009;296(4):L603–L13. pmid:19168577