Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 2655 2023-07-26 11:06:36 |
2 format change Meta information modification 2655 2023-07-26 11:26:12 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Zhu, H.; Chen, H.; Wen, H.; Wang, Z.; Liu, S. Engineered Lipidic Nanomaterials for Cancer Therapy. Encyclopedia. Available online: https://encyclopedia.pub/entry/47297 (accessed on 24 July 2024).
Zhu H, Chen H, Wen H, Wang Z, Liu S. Engineered Lipidic Nanomaterials for Cancer Therapy. Encyclopedia. Available at: https://encyclopedia.pub/entry/47297. Accessed July 24, 2024.
Zhu, Han, Hua-Jie Chen, Hai-Yan Wen, Zhi-Gang Wang, Shu-Lin Liu. "Engineered Lipidic Nanomaterials for Cancer Therapy" Encyclopedia, https://encyclopedia.pub/entry/47297 (accessed July 24, 2024).
Zhu, H., Chen, H., Wen, H., Wang, Z., & Liu, S. (2023, July 26). Engineered Lipidic Nanomaterials for Cancer Therapy. In Encyclopedia. https://encyclopedia.pub/entry/47297
Zhu, Han, et al. "Engineered Lipidic Nanomaterials for Cancer Therapy." Encyclopedia. Web. 26 July, 2023.
Engineered Lipidic Nanomaterials for Cancer Therapy
Edit

Sphingomyelin (SM) and its metabolites are crucial regulators of tumor cell growth, differentiation, senescence, and programmed cell death. With the rise in lipid-based nanomaterials, engineered lipidic nanomaterials inspired by SM metabolism, corresponding lipid targeting, and signaling activation have made fascinating advances in cancer therapeutic processes.

cancer therapy lipidic nanomaterial sphingomyelin metabolism tumor progression

1. Introduction

Human mortality is becoming increasingly dominated by cancer, which has long posed a significant public health concern [1]. According to the latest estimates by the World Health Organization (WHO)’s International Agency for Research on Cancer (IARC), the number of new cancer cases reported worldwide have risen to 19.29 million, with 9.96 million deaths reported by the year of 2020 [2]. For decades, scientists have been seeking effective cancer therapeutic targets and developing innovative cancer treatment strategies to defeat cancer. Over the past few decades, combining nanotechnology, materials science, and biotechnology has accelerated the development of the innovative nanomedicine field for cancer treatment [3]. Due to the unique appeal of nanomaterials for drug delivery, cancer diagnosis and imaging, and immune vaccine development, interest in applying nanotechnology to cancer treatment has grown, and a considerable level of technical success has been achieved [4][5]. Notable examples include small superparamagnetic iron oxide nanoparticles, which have been widely used as magnetic resonance imaging (MRI) contrast agents, and the successful introduction of the lipid nanoparticle COVID-19 mRNA vaccine [6][7]. Lipidic nanomaterials are nanoparticles consisting of lipid-like substances, with their diameters typically being between 10–200 nm [8]. As one of the most popular platforms for cancer treatment, they have demonstrated tremendous therapeutic potential across a wide range of treatment modalities. The natural origin of lipids ensures an ultra-high biocompatibility and degradability, making them superior to other materials like polymers due to their low probability of causing lipid-induced toxicity [8]. Moreover, they also exhibit attractive multifunctional properties, including an encapsulation capability for either hydrophilic or hydrophobic therapeutics, as well as surface properties, such as charge and targeting ligand modifications, that can be easily modified by altering the lipid composition or by attaching antibodies [9][10]. This flexibility allows for precise drug delivery and targeted therapy, making lipidic nanomaterials promising candidates for innovative cancer therapeutic strategies.
The emergence of lipidomic and lipid quantification in recent years has provided large-scale qualitative and quantitative studies of lipid compounds in organisms, providing insights into the functions and changes of lipid compounds under physiopathological conditions, and has also significantly contributed to the development of lipidic nanomaterials [11][12][13]. There is growing evidence in that when SM levels are expressed abnormally, it increases the cancer risk [14][15]. Among them, biological studies on the metabolism and function of sphingomyelin (SM) have revealed that effector molecules in the SM metabolic pathway contribute significantly to cell signaling, especially in regulating tumor cell growth, differentiation, senescence, and survival [16][17][18]. SM is integral for the membrane, and is an essential structural molecule in the cell membrane. Related enzymes, such as sphingomyelinase (SMase) and sphingomyelin synthase (SMS), strongly regulate its abundance [19]. A critical role for SM content and metabolic enzyme activity in signaling pathway transduction and therapy must be considered. Furthermore, SM metabolites have diverse biological functions across various cancerous processes [20]. For example, ceramide (Cer), produced by the SMase-mediated hydrolysis of SM, is closely associated with cell death, senescence, and cellular arrest [21][22]. Upon further metabolism, Cer can be converted to a pro-survival metabolite called sphingosine-1-phosphate (S1P), which is an anti-apoptotic molecule [23][24]. Thus, the dynamic transformation processes associated with SM metabolism are critical for mediating the fate of cancer cells.
Currently, an increasing number of investigators are focusing on the design of lipidic nanomaterials based on SM metabolism, initiating preclinical trials for different types of tumors to evaluate the potential application of therapeutic strategies utilizing SM, Cer, and S1P [25][26].

2. Synthesis and Metabolism of SM

2.1. Structure and Distribution of SM

In the 1880s, Johann Ludwig Wilhelm Thudichum [27] discovered and named the first sphingolipid SM, which is a phospholipid that is mainly found in vertebrates, accounting for 2% to 15% of total phospholipids, respectively [27]. Each molecule of SM is composed of a sphingosine (Sph) molecule, a fatty acid, and phosphorylcholine polar head group, and is characterized by a hydrophilic head and a hydrophobic tail made of two fatty acid chains, allowing them to form a lipid bilayer, the chemical structure of which is shown in Figure 1 (top left) [28][29]. The hydrophobic tail of SM is typically 16 to 24 carbons in length. At the same time, the sphingoid backbone can be either dihydrosphingosine, sphingosine, or 4-hydroxysphinganine, thus conferring heterogeneity to the SM molecule [30]. Various studies have shown that SM is present in most intracellular compartments, including the Golgi apparatus (which produces most of the SM molecules) and the plasma membrane (PM) (Figure 2) [16]. SM is also highly enriched in the outer leaflet of the PM and is the major component of the PM [31]. Exceptionally, erythrocytes and neural tissue have a higher SM content [20]. It has been estimated that the proportion of SM in the PM of fibroblasts can account for 40% to 90% of the total SM in the cell, respectively [32].
Figure 1. Chemical structure formula of sphingomyelin and its metabolites. The biological roles of the relevant signaling molecules are labeled under different colored fonts. Sphingomyelinase (SMase), sphingomyelin synthase (SMS), ceramidase (CDase), ceramide synthase (CerS), sphingosine kinase (SphK), and sphingosine-1-phosphate phosphatase (S1PP).
Figure 2. Sphingomyelin-related metabolic pathways. The relevant chemical structures are shown (above left). Ceramide (Cer) de novo synthesis starts from the endoplasmic reticulum (ER), and once synthesized, can be transferred to the trans-Golgi via the ceramide transfer protein (CERT), or be reorganized via a salvage pathway (red arrow). The action of sphingomyelin synthase 1 (SMS1) on Cer leads to the production of sphingomyelin (SM) (orange arrow). In response to the stimulus of apoptosis, phospholipid disorganization moves the isolated SM from the outer leaflet to the cytoplasmic side of the plasma membrane so that sphingomyelinase (SMase) can act on them to produce apoptotic Cer, with the opposite process occurring via sphingomyelin synthase (SMS) (black arrow). Second, Cer can be produced via the SMase (blue arrow). SM can also be produced by converting Cer via sphingomyelin synthase-associated protein (SMSr) (green arrow). Sphingosine (Sph) is produced by the hydrolysis of Cer by ceramidase (CDase), which is then phosphorylated by sphingosine kinase (SphK) to produce sphingosine-1-phosphate (S1P) (pink arrow). The presence of S1P lyase (SPL) also allows S1P to produce ethanolamine phosphate and hexadecenal (brown arrow).

2.2. SM Synthesis

SM is synthesized by two major isoforms of SMS, SMS1 (located in the trans-Golgi) and SMS2 (located in the trans-Golgi and PM) [33]. SMS2 in the PM is the only enzyme capable of synthesizing SM, and may have the unique function of directly maintaining SM content in the PM. It is unknown where the primary site of SM synthesis is located, but it occurs either on the luminal side of the trans-Golgi or in the extracellular leaflet of the PM (orange arrow in Figure 2) [34][35]. Cer is the substrate for this reaction, and this molecule is synthesized in the endoplasmic reticulum (ER). Thus, Cer must be transferred from its site of synthesis to the Golgi via a mediator termed the ceramide transfer protein (CERT) (a non-vesicular mechanism). The transfer of a phosphocholine headgroup from phosphatidylcholine to Cer to generate SM is catalyzed by SMS1, along with the generation of diacylglycerol (DAG) [36]. CERT is a cytoplasmic protein with two unique domains: the START (StAR-related lipid-transfer) domain for Cer recognition and the PH (pleckstrin homology) domain for phosphatidylinositol binding [36][37]. Unlike SMS1, SMS2 is not reliant on CERT-mediated Cer molecules to synthesize SM due to its unique cellular location, and is the only type of SMS present in the PM. Its most prominent mechanism is likely in converting local Cer to SM [38]. Sphingomyelin synthase-related protein (SMSr) is an enzyme present in the ER that is capable of converting Cer to ceramide phosphorylethanolamine (CPE), followed by the head group being exchanged or methylated to produce SM (green arrow in Figure 2) [31][39]. As CPE synthesizes extremely slowly, SM is produced in much lower amounts than when SM is synthesized in the Golgi, and it is commonly believed that SMSr regulates the balance of Cer in the ER to prevent excessive accumulation of Cer.

2.3. SM Metabolism

The metabolites of SM include Cer, Sph, and S1P, which are an essential class of biologically active signaling molecules [40]. Cer and Sph are important components of the stress response regulatory system, mediating cell cycle arrest and inducing apoptosis. However, S1P exhibits the opposite effect, promoting cell growth, invasion, and survival [41]. The synthesis and catabolism of these metabolites constitute a dynamic equilibrium relationship that judges the cell fates (e.g., cell proliferation, differentiation, or apoptosis). The regulation of these interconnected cell fates results from interactions and mutual equilibria between regulatory molecules within the cell. This subtle relationship between Cer, Sph, and S1P has been described as a “sphingomyelin rheostat” [21][22].

3. Role of SM in the Development of Tumors

Active molecules of the SM-related pathways play a crucial role in tumorigenesis and progression, and alterations in their related signaling play a vital role in the induction of cancer cell death or survival (Table 1).
Table 1. The role of active molecules in SM-related pathways in tumorigenesis and development.

Active Molecule

Pathway

Function or Activity

Cancer

Refs.

Sphingosine kinase

Akt/NF-κB

Cancer progression and chemoresistance

Colon

[42]

 

S1P/S1PR1

Inflammation and angiogenesis

Breast

[43]

 

PI3k/Akt/FOXO3a

Apoptosis resistance

Breast

[44]

 

S1P/Stat3/AKT

Proliferation

Colon

[45]

 

E-cad

Tumorigenesis and metastasis

Breast

[46]

 

S1P/ERK/CD44

Chemoresistance

Colon

[47]

 

S1P/AKT/GSK-3β

HIF-1α stabilization

Glioblastoma

[48]

Sphingomyelinase

 

Sensitivity

Glioblastoma

[49]

   

Resistance

Melanoma

[50]

   

Induce apoptosis

Colon

[51]

   

Induce apoptosis and resistance

Ovarian

[52]

Acid ceramidase

 

Proliferative Sensitivity

Melanoma

[53][54]

   

Sensitivity

Prostate

[55]

   

Radioresistant

Glioblastoma

[55]

Sphingosine kinase 2

Mcl-1

Cell survival

Leukemia

[56]

Sphingosine-1-phosphate lyase

p53 and p38

Apoptosis

Colon

[57]

Sphingomyelin synthase

TGF- b1

Migratory, invasion

Breast

[58]

 

Overexpression of SMS1

Cell death

Lymphatic

[59]

Sphingomyelinase

CD95

Apoptosis

Lymphatic

[60]

 

p53

Apoptosis

Lung

[61]

Sphingosine

Cdk4

Cell proliferation

Intestinal adenoma

[62]

Ceramide

CerS6/C16-ceramide activated

Apoptosis

Lung

[63]

 

High cytotoxicity in p53

Apoptosis

Breast

[64]

↑, increase; ↓, decrease.

4. SM Metabolism-Based Lipidic Nanomaterials for Cancer Therapy

4.1. SM-Based Lipidic Nanomaterials for Cancer Therapy

Various chemotherapeutic agents, small molecule inhibitors, and microRNAs (miRNAs) have all been used for cancer treatment in the clinic, but often have a poor therapeutic efficacy due to their low in vivo utilization and off-target [65]. To improve their clinical translation, various nanocarriers have been designed to achieve precise targeting and low side effects, enhanced permeability, and retention effects [66]. SM nanosystems have been shown to be promising carriers for effectively delivering anticancer drugs. Recently, Wang et al. [67] developed an SM-derived camptothecin (CPT) (SM-CPT) liposome nanotherapeutic platform, in which the hydroxyl group of SM is partially suffixed with CPT in self-assembly, driven by the amphiphilic nature of SM in aqueous media. This enhances lactone stability and triggers a cytotoxic T lymphocyte (CTL) response to activating anti-tumor immunity. Coupling this with a PD-L1/PD-1 blockade eradicated more than 80% of MC38 colon tumors. Based on the excellent ability of SM nanosystems to bind to different types of drugs, Bouzo et al. [68] proposed the chemical modification of the natural ligands of guanylyl cyclase (GCC) receptors expressed in metastatic colorectal cancer tumors, which were then anchored to SM nanosystems, and finally loaded with the anticancer drug etoposide (Etp) for combination therapy, a strategy that has previously shown strong potential for the treatment of metastatic colorectal cancer cells. Medina et al. [69] increased the homing ability of pancreatic ductal adenocarcinoma (PDAC) tumors for pancreatic tumor imaging by encapsulating iron nanoparticles and the indocyanine green (ICG) in liposomes composed of cationic SM, while embedding RA-96 Fab fragments at the surface of the liposomal nanoparticles. This may be a suitable drug delivery tool for enhancing positive therapeutic outcomes in PDAC patients. Another novel active release system is the encapsulation of cisplatin and ICG in magnetic SM liposomes for their release in response to stress cellular SMase. Individual iron particles and large clusters of iron particle structural domains can be observed in TEM images upon iron and SMase treatment and are clearly distinguishable from the untreated control liposomes. Intracellular release studies were subsequently performed with Texas red membrane-labeled liposomes loaded with calreticulin, and CLSM images demonstrated a high diffusion of calreticulin under alternating magnetic field (AMF) treatment. This design allowed the system to only open at the appropriate time within the lesion, showing an improved therapeutic efficacy in murine squamous cell carcinoma tumors [70]. RNA-based gene therapy is a prospective option in the fight against cancer, but the efficiency of biological delivery requires further optimization. Farimah et al. [71] developed a gene therapy nanodrug capable of interfering with tumor growth and migration using a biocompatible SM nanosystem to effectively deliver encoded TP53TG1 plasmids to HCT-116 colorectal cells as treatment. Another SM-based nanosystem (SNs) incorporating a cationic lipid stearylamine (ST) has been developed that is able to support miRNA conjugation by establishing electrostatic interactions (SNs–ST) [72].

4.2. Cer-Based Lipidic Nanomaterials for Cancer Therapy

Cer-mediated anticancer effects have been demonstrated in various cancers (pancreatic, breast, and gastric), affecting T-cell signaling and inducing apoptosis in cancer cells [73][74][75]. However, the cellular impermeability and hydrophobicity of Cer greatly limits its practical application. Several delivery vehicles have been developed to increase Cer delivery and enhance its pharmacological effects, such as calcium phosphate nanocomposite particles, nanoemulsions, liposomes, and others [76][77][78][79]. Li et al. [80] developed C6-Cer-containing nanoliposomes (LipC6) to overcome this limitation for improving the efficacy of immunotherapy in patients with liver cancer.

4.3. S1P-Based Lipidic Nanomaterials for Cancer Therapy

S1P is a lipid member of the SM metabolic pathway that facilitates tumor progression. To address the lack of effective targets for GBM and the low accumulation of drugs in the diseased area from the blood–brain-tumor barrier (BBTB), Liu et al. [81] developed a liposome-based GBM drug delivery system, S1P/JS-K/Lipo, which utilized S1P as its active lipid ligand. This enabled the active penetration of nitric oxide (NO) prodrugs (JS-K, O2-(2,4-dinitrophenyl) 1-[(4-ethoxycarbonyl) piperazin-1-yl] diazen-1-ium-1,2-diolate) into the BBTB to produce tiny NO bubbles for killing tumor cells. When S1P/JS-K/Lipo (equivalent to 2 × 10−3 mg kg−1 body weight of S1P and 1 mg kg−1 body weight of JS-K) was intravenously administered to mice, significant inhibitions of U87MG-RFP-tumor growth was observed. The system was also demonstrated to adequately interact specifically with the highly expressed S1PRs on GBM cells for an efficient targeted delivery. The FDA-approved drug FTY720 (fingolimod) impairs acid-induced osteosarcoma cell survival and migration by reducing S1P levels. It is currently in the experimental phase as an immunomodulatory agent, and may be useful in designing nanotherapeutic agents [82]. To date, various types of S1P signaling modulators have been developed, including S1P agonists, S1P antagonists, and SphK inhibitors [83]. Their positive impact on cancer therapy (including, but not limited to increased sensitivity to chemotherapy and improved oncologic outcomes) has been described in the literature [84][85][86]. Among them includes the sphingolipid phosphatase SGPP1, an antagonist of S1P signaling which can improve the radiosensitivity of miR-95 overexpressed mice [87]. Additionally, restraining S1P synthesis by inhibiting SphK activity can be used to heighten the response of cancer cells to cytotoxic treatments, and these studies will be a strong basis for future nanomedicine design [88][89].
Finally, recent lipidic nanomaterials designed based on SM metabolism for cancer therapy were summarized, and their related parameters are listed in Table 2.
Table 2. SM metabolism-based lipidic nanomaterials for cancer therapy.

References

  1. Miller, K.D.; Siegel, R.L.; Lin, C.C.; Mariotto, A.B.; Kramer, J.L.; Rowland, J.H.; Stein, K.D.; Alteri, R.; Jemal, A. Cancer treatment and survivorship statistics, 2016. CA Cancer J. Clin. 2016, 66, 271–289.
  2. Ferlay, J.; Colombet, M.; Soerjomataram, I.; Parkin, D.M.; Pineros, M.; Znaor, A.; Bray, F. Cancer statistics for the year 2020: An overview. Int. J. Cancer 2021, 149, 778–789.
  3. Riehemann, K.; Schneider, S.W.; Luger, T.A.; Godin, B.; Ferrari, M.; Fuchs, H. Nanomedicine--challenge and perspectives. Angew. Chem. Int. Ed. 2009, 48, 872–897.
  4. Zhou, Q.; Zhang, L.; Wu, H. Nanomaterials for cancer therapies. Nanotechnol. Rev. 2017, 6, 473–496.
  5. Sun, T.; Zhang, Y.S.; Pang, B.; Hyun, D.C.; Yang, M.; Xia, Y. Engineered nanoparticles for drug delivery in cancer therapy. Angew. Chem. Int. Ed. 2014, 53, 12320–12364.
  6. Dadfar, S.M.; Camozzi, D.; Darguzyte, M.; Roemhild, K.; Varvara, P.; Metselaar, J.; Banala, S.; Straub, M.; Guvener, N.; Engelmann, U.; et al. Size-isolation of superparamagnetic iron oxide nanoparticles improves MRI, MPI and hyperthermia performance. J. Nanobiotechnol. 2020, 18, 22.
  7. Li, M.; Huang, Y.; Wu, J.; Li, S.; Mei, M.; Chen, H.; Wang, N.; Wu, W.; Zhou, B.; Tan, X.; et al. A PEG-lipid-free COVID-19 mRNA vaccine triggers robust immune responses in mice. Mater. Horiz. 2023, 10, 466–472.
  8. Chaudhari, V.S.; Murty, U.S.; Banerjee, S. Lipidic nanomaterials to deliver natural compounds against cancer: A review. Environ. Chem. Lett. 2020, 18, 1803–1812.
  9. Limongi, T.; Susa, F.; Marini, M.; Allione, M.; Torre, B.; Pisano, R.; di Fabrizio, E. Lipid-based nanovesicular drug delivery systems. Nanomaterials 2021, 11, 3391.
  10. Porter, C.J.; Trevaskis, N.L.; Charman, W.N. Lipids and lipid-based formulations: Optimizing the oral delivery of lipophilic drugs. Nat. Rev. Drug Discov. 2007, 6, 231–248.
  11. Quehenberger, O.; Armando, A.M.; Brown, A.H.; Milne, S.B.; Myers, D.S.; Merrill, A.H.; Bandyopadhyay, S.; Jones, K.N.; Kelly, S.; Shaner, R.L.; et al. Lipidomics reveals a remarkable diversity of lipids in human plasma. J. Lipid Res. 2010, 51, 3299–3305.
  12. Wolrab, D.; Jirasko, R.; Cifkova, E.; Horing, M.; Mei, D.; Chocholouskova, M.; Peterka, O.; Idkowiak, J.; Hrnciarova, T.; Kuchar, L.; et al. Lipidomic profiling of human serum enables detection of pancreatic cancer. Nat. Commun. 2022, 13, 124.
  13. Hu, Y.; Zhang, R.Q.; Wang, Z.G.; Liu, S.L. In situ quantification of lipids in live cells by using lipid-binding domain-based biosensors. Bioconjug. Chem. 2022, 33, 2076–2087.
  14. Radin, N.S. Cancer progression in the kidney and prostate: Vital roles of sphingolipids in chemotherapy. Urology 2002, 60, 562–568.
  15. Vykoukal, J.; Fahrmann, J.F.; Gregg, J.R.; Tang, Z.; Basourakos, S.; Irajizad, E.; Park, S.; Yang, G.; Creighton, C.J.; Fleury, A.; et al. Caveolin-1-mediated sphingolipid oncometabolism underlies a metabolic vulnerability of prostate cancer. Nat. Commun. 2020, 11, 4279.
  16. D’Angelo, G.; Moorthi, S.; Luberto, C. Role and function of sphingomyelin biosynthesis in the development of cancer. Adv. Cancer Res. 2018, 140, 61–96.
  17. Bienias, K.; Fiedorowicz, A.; Sadowska, A.; Prokopiuk, S.; Car, H. Regulation of sphingomyelin metabolism. Pharmacol. Rep. 2016, 68, 570–581.
  18. Ogretmen, B. Sphingolipid metabolism in cancer signalling and therapy. Nat. Rev. Cancer 2018, 18, 33–50.
  19. Ogretmen, B.; Hannun, Y.A. Biologically active sphingolipids in cancer pathogenesis and treatment. Nat. Rev. Cancer 2004, 4, 604–616.
  20. Modrak, D.E.; Gold, D.V.; Goldenberg, D.M. Sphingolipid targets in cancer therapy. Mol. Cancer Ther. 2006, 5, 200–208.
  21. Newton, J.; Lima, S.; Maceyka, M.; Spiegel, S. Revisiting the sphingolipid rheostat: Evolving concepts in cancer therapy. Exp. Cell Res. 2015, 333, 195–200.
  22. Taniguchi, M.; Okazaki, T. Role of ceramide/sphingomyelin (SM) balance regulated through “SM cycle” in cancer. Cell. Signal. 2021, 87, 110119.
  23. Cartier, A.; Hla, T. Sphingosine 1-phosphate: Lipid signaling in pathology and therapy. Science 2019, 366, eaar5551.
  24. Taha, T.A.; Mullen, T.D.; Obeid, L.M. A house divided: Ceramide, sphingosine, and sphingosine-1-phosphate in programmed cell death. Biochim. Biophys. Acta 2006, 1758, 2027–2036.
  25. Kolesnick, R. The therapeutic potential of modulating the ceramide/sphingomyelin pathway. J. Clin. Investig. 2002, 110, 3–8.
  26. Huwiler, A.; Pfeilschifter, J. Altering the sphingosine-1-phosphate/ceramide balance: A promising approach for tumor therapy. Curr. Pharm. Des. 2006, 12, 4625–4635.
  27. Merrill, A.H., Jr.; Schmelz, E.M.; Dillehay, D.L.; Spiegel, S.; Shayman, J.A.; Schroeder, J.J.; Riley, R.T.; Voss, K.A.; Wang, E. Sphingolipids--the enigmatic lipid class: Biochemistry, physiology, and pathophysiology. Toxicol. Appl. Pharmacol. 1997, 142, 208–225.
  28. Filippov, A.; Oradd, G.; Lindblom, G. Sphingomyelin structure influences the lateral diffusion and raft formation in lipid bilayers. Biophys. J. 2006, 90, 2086–2092.
  29. Christie; William, W. Lipids: Their structures and occurrence. In Lipid Analysis, 4th ed.; Elsevier: Amsterdam, The Netherlands, 2012; pp. 3–19.
  30. Furland, N.E.; Zanetti, S.R.; Oresti, G.M.; Maldonado, E.N.; Aveldano, M.I. Ceramides and sphingomyelins with high proportions of very long-chain polyunsaturated fatty acids in mammalian germ cells. J. Biol. Chem. 2007, 282, 18141–18150.
  31. Slotte, J.P. Biological functions of sphingomyelins. Prog. Lipid Res. 2013, 52, 424–437.
  32. Allan, D.; Quinn, P. Resynthesis of sphingomyelin from plasma-membrane phosphatidylcholine in BHK cells treated with staphylococcus aureus sphingomyelinase. Biochem. J. 1988, 254, 765–771.
  33. Gault, C.R.; Obeid, L.M.; Hannun, Y.A. An overview of sphingolipid metabolism: From synthesis to breakdown. Adv. Exp. Med. Biol. 2010, 688, 1–23.
  34. Futerman, A.H.; Stieger, B.; Hubbard, A.L.; Pagano, R.E. Sphingomyelin synthesis in rat liver occurs predominantly at the cis and medial cisternae of the Golgi apparatus. J. Biol. Chem. 1990, 265, 8650–8657.
  35. Huitema, K.; van den Dikkenberg, J.; Brouwers, J.F.; Holthuis, J.C. Identification of a family of animal sphingomyelin synthases. EMBO J. 2004, 23, 33–44.
  36. Hanada, K.; Kumagai, K.; Yasuda, S.; Miura, Y.; Kawano, M.; Fukasawa, M.; Nishijima, M. Molecular machinery for non-vesicular trafficking of ceramide. Nature 2003, 426, 803–809.
  37. Hanada, K. Regulation of CERT-mediated trafficking of ceramide. Chem. Phys. Lipids 2007, 149, S7.
  38. Mitsutake, S.; Zama, K.; Yokota, H.; Yoshida, T.; Tanaka, M.; Mitsui, M.; Ikawa, M.; Okabe, M.; Tanaka, Y.; Yamashita, T.; et al. Dynamic modification of sphingomyelin in lipid microdomains controls development of obesity, fatty liver, and type 2 diabetes. J. Biol. Chem. 2011, 286, 28544–28555.
  39. Vacaru, A.M.; Tafesse, F.G.; Ternes, P.; Kondylis, V.; Hermansson, M.; Brouwers, J.F.; Somerharju, P.; Rabouille, C.; Holthuis, J.C. Sphingomyelin synthase-related protein SMSr controls ceramide homeostasis in the ER. J. Cell Biol. 2009, 185, 1013–1027.
  40. Saddoughi, S.A.; Song, P.; Ogretmen, B. Roles of bioactive sphingolipids in cancer biology and therapeutics. Subcell. Biochem. 2008, 49, 413–440.
  41. Haddadi, N.; Lin, Y.; Simpson, A.M.; Nassif, N.T.; McGowan, E.M. “Dicing and splicing” sphingosine kinase and relevance to cancer. Int. J. Mol. Sci. 2017, 18, 1891.
  42. Shamekhi, S.; Abdolalizadeh, J.; Ostadrahimi, A.; Mohammadi, S.A.; Barzegari, A.; Lotfi, H.; Bonabi, E.; Zarghami, N. Apoptotic effect of saccharomyces cerevisiae on human colon cancer SW480 cells by regulation of Akt/NF-kB signaling pathway. Probiotics Antimicrob. Proteins 2020, 12, 311–319.
  43. Nagahashi, M.; Yamada, A.; Katsuta, E.; Aoyagi, T.; Huang, W.C.; Terracina, K.P.; Hait, N.C.; Allegood, J.C.; Tsuchida, J.; Yuza, K.; et al. Targeting the SphK1/S1P/S1PR1 axis that links obesity, chronic inflammation, and breast cancer metastasis. Cancer Res. 2018, 78, 1713–1725.
  44. Zhang, X.; Zhuang, T.; Liang, Z.; Li, L.; Xue, M.; Liu, J.; Liang, H. Breast cancer suppression by aplysin is associated with inhibition of PI3K/AKT/FOXO3a pathway. Oncotarget 2017, 8, 63923–63934.
  45. Bao, Y.; Li, K.; Guo, Y.; Wang, Q.; Li, Z.; Yang, Y.; Chen, Z.; Wang, J.; Zhao, W.; Zhang, H.; et al. Tumor suppressor PRSS8 targets Sphk1/S1P/Stat3/Akt signaling in colorectal cancer. Oncotarget 2016, 7, 26780–26792.
  46. Zheng, X.D.; Zhang, Y.; Qi, X.W.; Wang, M.H.; Sun, P.; Zhang, Y.; Jiang, J. Role of Sphk1 in the malignant transformation of breast epithelial cells and breast cancer progression. Indian J. Cancer 2014, 51, 524–529.
  47. Kawahara, S.; Otsuji, Y.; Nakamura, M.; Murakami, M.; Murate, T.; Matsunaga, T.; Kanoh, H.; Seishima, M.; Banno, Y.; Hara, A. Sphingosine kinase 1 plays a role in the upregulation of CD44 expression through extracellular signal-regulated kinase signaling in human colon cancer cells. Anticancer Drugs 2013, 24, 473–483.
  48. Ader, I.; Brizuela, L.; Bouquerel, P.; Malavaud, B.; Cuvillier, O. Sphingosine kinase 1: A new modulator of hypoxia inducible factor 1alpha during hypoxia in human cancer cells. Cancer Res. 2008, 68, 8635–8642.
  49. Pchejetski, D.; Golzio, M.; Bonhoure, E.; Calvet, C.; Doumerc, N.; Garcia, V.; Mazerolles, C.; Rischmann, P.; Teissie, J.; Malavaud, B.; et al. Sphingosine kinase-1 as a chemotherapy sensor in prostate adenocarcinoma cell and mouse models. Cancer Res. 2005, 65, 11667–11675.
  50. Alshaker, H.; Wang, Q.; Kawano, Y.; Arafat, T.; Bohler, T.; Winkler, M.; Cooper, C.; Pchejetski, D. Everolimus (RAD001) sensitizes prostate cancer cells to docetaxel by down-regulation of HIF-1alpha and sphingosine kinase 1. Oncotarget 2016, 7, 80943–80956.
  51. Sauer, L.; Nunes, J.; Salunkhe, V.; Skalska, L.; Kohama, T.; Cuvillier, O.; Waxman, J.; Pchejetski, D. Sphingosine kinase 1 inhibition sensitizes hormone-resistant prostate cancer to docetaxel. Int. J. Cancer 2009, 125, 2728–2736.
  52. Sukocheva, O.; Wang, L.; Verrier, E.; Vadas, M.A.; Xia, P. Restoring endocrine response in breast cancer cells by inhibition of the sphingosine kinase-1 signaling pathway. Endocrinology 2009, 150, 4484–4492.
  53. Giussani, P.; Bassi, R.; Anelli, V.; Brioschi, L.; De Zen, F.; Riccitelli, E.; Caroli, M.; Campanella, R.; Gaini, S.M.; Viani, P.; et al. Glucosylceramide synthase protects glioblastoma cells against autophagic and apoptotic death induced by temozolomide and Paclitaxel. Cancer Investig. 2012, 30, 27–37.
  54. Liu, Y.Y.; Han, T.Y.; Giuliano, A.E.; Cabot, M.C. Expression of glucosylceramide synthase, converting ceramide to glucosylceramide, confers adriamycin resistance in human breast cancer cells. J. Biol. Chem. 1999, 274, 1140–1146.
  55. Liu, Y.Y.; Patwardhan, G.A.; Xie, P.; Gu, X.; Giuliano, A.E.; Cabot, M.C. Glucosylceramide synthase, a factor in modulating drug resistance, is overexpressed in metastatic breast carcinoma. Int. J. Oncol. 2011, 39, 425–431.
  56. LeBlanc, F.R.; Pearson, J.M.; Tan, S.F.; Cheon, H.; Xing, J.C.; Dunton, W.; Feith, D.J.; Loughran, T.P., Jr. Sphingosine kinase-2 is overexpressed in large granular lymphocyte leukaemia and promotes survival through Mcl-1. Br. J. Heaematol. 2020, 190, 405–417.
  57. Oskouian, B.; Sooriyakumaran, P.; Borowsky, A.D.; Crans, A.; Dillard-Telm, L.; Tam, Y.Y.; Bandhuvula, P.; Saba, J.D. Sphingosine-1-phosphate lyase potentiates apoptosis via p53- and p38-dependent pathways and is down-regulated in colon cancer. Proc. Natl. Acad. Sci. USA 2006, 103, 17384–17389.
  58. Zheng, K.; Chen, Z.; Feng, H.; Chen, Y.; Zhang, C.; Yu, J.; Luo, Y.; Zhao, L.; Jiang, X.; Shi, F. Sphingomyelin synthase 2 promotes an aggressive breast cancer phenotype by disrupting the homoeostasis of ceramide and sphingomyelin. Cell Death Dis. 2019, 10, 157.
  59. Lafont, E.; Milhas, D.; Carpentier, S.; Garcia, V.; Jin, Z.X.; Umehara, H.; Okazaki, T.; Schulze-Osthoff, K.; Levade, T.; Benoist, H.; et al. Caspase-mediated inhibition of sphingomyelin synthesis is involved in FasL-triggered cell death. Cell Death Differ. 2010, 17, 642–654.
  60. Grassme, H.; Schwarz, H.; Gulbins, E. Molecular mechanisms of ceramide-mediated CD95 clustering. Biochem. Biophys. Res. Commun. 2001, 284, 1016–1030.
  61. Eroica, S.; Susan, C.E.; Cynthia, C.; Elroy, F. Characterizing the sphingomyelinase pathway triggered by PRIMA-1 derivatives in lung cancer cells with differing p53 status. Anticancer Res. 2014, 34, 3271.
  62. Kohno, M.; Momoi, M.; Oo, M.L.; Paik, J.H.; Lee, Y.M.; Venkataraman, K.; Ai, Y.; Ristimaki, A.P.; Fyrst, H.; Sano, H.; et al. Intracellular role for sphingosine kinase 1 in intestinal adenoma cell proliferation. Mol. Cell. Biol. 2006, 26, 7211–7223.
  63. Senkal, C.E.; Ponnusamy, S.; Manevich, Y.; Meyers-Needham, M.; Saddoughi, S.A.; Mukhopadyay, A.; Dent, P.; Bielawski, J.; Ogretmen, B. Alteration of ceramide synthase 6/C16-ceramide induces activating transcription factor 6-mediated endoplasmic reticulum (ER) stress and apoptosis via perturbation of cellular Ca2+ and ER/Golgi membrane network. J. Biol. Chem. 2011, 286, 42446–42458.
  64. Chang, W.T.; Wu, C.Y.; Lin, Y.C.; Wu, M.T.; Su, K.L.; Yuan, S.S.; Wang, H.D.; Fong, Y.; Lin, Y.H.; Chiu, C.C. C(2)-ceramide-induced rb-dominant senescence-like phenotype leads to human breast cancer Mcf-7 escape from p53-dependent cell death. Int. J. Mol. Sci. 2019, 20, 4292.
  65. Zhong, L.; Li, Y.; Xiong, L.; Wang, W.; Wu, M.; Yuan, T.; Yang, W.; Tian, C.; Miao, Z.; Wang, T.; et al. Small molecules in targeted cancer therapy: Advances, challenges, and future perspectives. Signal Transduct. Target. Ther. 2021, 6, 201.
  66. Phillips, M.A.; Gran, M.L.; Peppas, N.A. Targeted nanodelivery of drugs and diagnostics. Nano Today 2010, 5, 143–159.
  67. Wang, Z.; Little, N.; Chen, J.; Lambesis, K.T.; Le, K.T.; Han, W.; Scott, A.J.; Lu, J. Immunogenic camptothesome nanovesicles comprising sphingomyelin-derived camptothecin bilayers for safe and synergistic cancer immunochemotherapy. Nat. Nanotechnol. 2021, 16, 1130–1140.
  68. Bouzo, B.L.; Lores, S.; Jatal, R.; Alijas, S.; Alonso, M.J.; Conejos-Sanchez, I.; de la Fuente, M. Sphingomyelin nanosystems loaded with uroguanylin and etoposide for treating metastatic colorectal cancer. Sci. Rep. 2021, 11, 17213.
  69. Medina, O.P.; Tower, R.J.; Medina, T.P.; Ashkenani, F.; Appold, L.; Bötcher, M.; Huber, L.; Will, O.; Ling, Q.; Hauser, C.; et al. Multimodal targeted nanoparticle-based delivery system for pancreatic tumor imaging in cellular and animal models. Curr. Pharm. Des. 2020, 28, 313–323.
  70. Penate Medina, T.; Gerle, M.; Humbert, J.; Chu, H.; Kopnick, A.L.; Barkmann, R.; Garamus, V.M.; Sanz, B.; Purcz, N.; Will, O.; et al. Lipid-iron nanoparticle with a cell stress release mechanism combined with a local alternating magnetic field enables site-activated drug release. Cancers 2020, 12, 3767.
  71. Masoumi, F.; Saraiva, S.M.; Bouzo, B.L.; Lopez-Lopez, R.; Esteller, M.; Diaz-Lagares, A.; de la Fuente, M. Modulation of colorectal tumor behavior via lncrna tp53tg1-lipidic nanosystem. Pharmaceutics 2021, 13, 1507.
  72. Nagachinta, S.; Bouzo, B.L.; Vazquez-Rios, A.J.; Lopez, R.; Fuente, M. Sphingomyelin-based nanosystems (sns) for the development of anticancer miRNA therapeutics. Pharmaceutics 2020, 12, 189.
  73. Morad, S.A.; Messner, M.C.; Levin, J.C.; Abdelmageed, N.; Park, H.; Merrill, A.H., Jr.; Cabot, M.C. Potential role of acid ceramidase in conversion of cytostatic to cytotoxic end-point in pancreatic cancer cells. Cancer Chemoth. Pharm. 2013, 71, 635–645.
  74. Flowers, M.; Fabrias, G.; Delgado, A.; Casas, J.; Abad, J.L.; Cabot, M.C. C6-ceramide and targeted inhibition of acid ceramidase induce synergistic decreases in breast cancer cell growth. Breast Cancer Res. Treat. 2012, 133, 447–458.
  75. Huang, H.; Zhang, Y.; Liu, X.; Li, Z.; Xu, W.; He, S.; Huang, Y.; Zhang, H. Acid sphingomyelinase contributes to evodiamine-induced apoptosis in human gastric cancer SGC-7901 cells. DNA Cell Biol. 2011, 30, 407–412.
  76. Kester, M.; Heakal, Y.; Fox, T.; Sharma, A.; Robertson, G.P.; Morgan, T.T.; Altinoglu, E.I.; Tabakovic, A.; Parette, M.R.; Rouse, S.M.; et al. Calcium phosphate nanocomposite particles for in vitro imaging and encapsulated chemotherapeutic drug delivery to cancer cells. Nano Lett. 2008, 8, 4116–4121.
  77. Ganta, S.; Singh, A.; Patel, N.R.; Cacaccio, J.; Rawal, Y.H.; Davis, B.J.; Amiji, M.M.; Coleman, T.P. Development of EGFR-targeted nanoemulsion for imaging and novel platinum therapy of ovarian cancer. Pharm. Res. 2014, 31, 2490–2502.
  78. Stover, T.; Kester, M. Liposomal delivery enhances short-chain ceramide-induced apoptosis of breast cancer cells. J. Pharmacol. Exp. Ther. 2003, 307, 468–475.
  79. Chen, L.; Alrbyawi, H.; Poudel, I.; Arnold, R.D.; Babu, R.J. Co-delivery of doxorubicin and ceramide in a liposomal formulation enhances cytotoxicity in murine B16Bl6 melanoma cell lines. AAPS PharmSciTech 2019, 20, 99.
  80. Li, G.; Liu, D.; Kimchi, E.T.; Kaifi, J.T.; Qi, X.; Manjunath, Y.; Liu, X.; Deering, T.; Avella, D.M.; Fox, T.; et al. Nanoliposome C6-ceramide increases the anti-tumor immune response and slows growth of liver tumors in mice. Gastroenterology 2018, 154, 1024–1036.
  81. Liu, Y.; Wang, X.; Li, J.; Tang, J.; Li, B.; Zhang, Y.; Gu, N.; Yang, F. Sphingosine 1-phosphate liposomes for targeted nitric oxide delivery to mediate anticancer effects against brain glioma tumors. Adv. Mater. 2021, 33, e2101701.
  82. Saddoughi, S.A.; Gencer, S.; Peterson, Y.K.; Ward, K.E.; Mukhopadhyay, A.; Oaks, J.; Bielawski, J.; Szulc, Z.M.; Thomas, R.J.; Selvam, S.P.; et al. Sphingosine analogue drug FTY720 targets I2PP2A/SET and mediates lung tumour suppression via activation of PP2A-RIPK1-dependent necroptosis. EMBO Mol. Med. 2013, 5, 105–121.
  83. Companioni, O.; Mir, C.; Garcia-Mayea, Y.; ME, L.L. Targeting Sphingolipids for Cancer Therapy. Front. Oncol. 2021, 11, 745092.
  84. Meng, Q.; Zhao, B.; Xu, Q.; Xu, X.; Deng, G.; Li, C.; Luan, L.; Ren, F.; Wang, H.; Xu, H. Indole-propionic acid derivatives as potent, S1P3-sparing and EAE efficacious sphingosine-1-phosphate 1 (S1P1) receptor agonists. Bioorg. Med. Chem. Lett. 2012, 22, 2794–2797.
  85. Luo, D.; Guo, Z.; Zhao, X.; Wu, L.; Liu, X.; Zhang, Y.; Zhang, Y.; Deng, Z.; Qu, X.; Cui, S.; et al. Novel 5-fluorouracil sensitizers for colorectal cancer therapy: Design and synthesis of S1P receptor 2 (S1PR2) antagonists. Eur. J. Med. Chem. 2021, 227, 113923.
  86. Beljanski, V.; Knaak, C.; Smith, C.D. A novel sphingosine kinase inhibitor induces autophagy in tumor cells. J. Pharmacol. Exp. Ther. 2010, 333, 454–464.
  87. Huang, X.; Taeb, S.; Jahangiri, S.; Emmenegger, U.; Tran, E.; Bruce, J.; Mesci, A.; Korpela, E.; Vesprini, D.; Wong, C.S.; et al. miRNA-95 mediates radioresistance in tumors by targeting the sphingolipid phosphatase SGPP1. Cancer Res. 2013, 73, 6972–6986.
  88. Billich, A.; Bornancin, F.; Mechtcheriakova, D.; Natt, F.; Huesken, D.; Baumruker, T. Basal and induced sphingosine kinase 1 activity in A549 carcinoma cells: Function in cell survival and IL-1beta and TNF-alpha induced production of inflammatory mediators. Cell. Signal. 2005, 17, 1203–1217.
  89. Leroux, M.E.; Auzenne, E.; Evans, R.; Hail, N., Jr.; Spohn, W.; Ghosh, S.C.; Farquhar, D.; McDonnell, T.; Klostergaard, J. Sphingolipids and the sphingosine kinase inhibitor, SKI II, induce BCL-2-independent apoptosis in human prostatic adenocarcinoma cells. Prostate 2007, 67, 1699–1717.
  90. Liu, H.; Kong, Y.; Liu, Z.; Guo, X.; Yang, B.; Yin, T.; He, H.; Gou, J.; Zhang, Y.; Tang, X. Sphingomyelin-based PEGylation Cu (DDC)(2) liposomes prepared via the dual function of Cu(2+) for cancer therapy: Facilitating DDC loading and exerting synergistic antitumor effects. Int. J. Pharm. 2022, 621, 121788.
  91. Bidan, N.; Lores, S.; Vanhecke, A.; Nicolas, V.; Domenichini, S.; Lopez, R.; de la Fuente, M.; Mura, S. Before in vivo studies: In vitro screening of sphingomyelin nanosystems using a relevant 3D multicellular pancreatic tumor spheroid model. Int. J. Pharm. 2022, 617, 121577.
  92. Massiot, J.; Rosilio, V.; Ibrahim, N.; Yamamoto, A.; Nicolas, V.; Konovalov, O.; Tanaka, M.; Makky, A. Newly synthesized lipid-porphyrin conjugates: Evaluation of their self-assembling properties, their miscibility with phospholipids and their photodynamic activity in vitro. Chemistry 2018, 24, 19179–19194.
  93. Lim, E.B.; Haam, S.; Lee, S.W. Sphingomyelin-based liposomes with different cholesterol contents and polydopamine coating as a controlled delivery system. Colloid Surface A 2021, 618, 126447.
  94. Zembruski, N.C.; Nguyen, C.D.; Theile, D.; Ali, R.M.; Herzog, M.; Hofhaus, G.; Heintz, U.; Burhenne, J.; Haefeli, W.E.; Weiss, J. Liposomal sphingomyelin influences the cellular lipid profile of human lymphoblastic leukemia cells without effect on P-glycoprotein activity. Mol. Pharm. 2013, 10, 1020–1034.
  95. Teng, Y.; Li, J.; Hui, S. C6 ceramide potentiates curcumin-induced cell death and apoptosis in melanoma cell lines in vitro. Cancer Chemother. Pharmacol. 2010, 66, 999–1003.
  96. Pal, S.; Medatwal, N.; Kumar, S.; Kar, A.; Komalla, V.; Yavvari, P.S.; Mishra, D.; Rizvi, Z.A.; Nandan, S.; Malakar, D.; et al. A localized chimeric hydrogel therapy combats tumor progression through alteration of sphingolipid metabolism. ACS Cent. Sci. 2019, 5, 1648–1662.
  97. Medatwal, N.; Ansari, M.N.; Kumar, S.; Pal, S.; Jha, S.K.; Verma, P.; Rana, K.; Dasgupta, U.; Bajaj, A. Hydrogel-mediated delivery of celastrol and doxorubicin induces a synergistic effect on tumor regression via upregulation of ceramides. Nanoscale 2020, 12, 18463–18475.
  98. Zhang, X.; Kitatani, K.; Toyoshima, M.; Ishibashi, M.; Usui, T.; Minato, J.; Egiz, M.; Shigeta, S.; Fox, T.; Deering, T.; et al. Ceramide nanoliposomes as a MLKL-dependent, necroptosis-inducing, chemotherapeutic reagent in ovarian cancer. Mol. Cancer Ther. 2018, 17, 50–59.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , ,
View Times: 362
Revisions: 2 times (View History)
Update Date: 26 Jul 2023
1000/1000
Video Production Service