Skip to main content
Erschienen in: Clinical and Translational Oncology 11/2022

Open Access 27.07.2022 | Review Article

The role of liver kinase B1 in tumor progression through regulation of lipid metabolism

verfasst von: Jialu Geng, Yanghe Zhang, Qingfei Meng, Hang Yan, Yishu Wang

Erschienen in: Clinical and Translational Oncology | Ausgabe 11/2022

Abstract

The somatic mutation of liver kinase B1 (LKB1) has been implicated in various tumors, which is reflected in the survival, proliferation, and metastasis of tumor cells. However, the regulation of LKB1 in lipid metabolism, a process that is involved in tumor progression is not completely clear. We conclude that LKB1 deficiency results in abnormal expression and activation of multiple molecules related to lipid metabolism which locate downstream of AMP-activated protein kinase (AMPK) or salt-induced kinase (SIK). Abnormal lipid metabolism induced by LKB1 deficiency contributes to the proliferation and metastasis of tumor cells through energy regulation.
Hinweise
Jialu Geng and Yanghe Zhang contributed equally to this work.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
LKB1
Liver kinase B1
STK
Serine-threonine kinase
HCC
Hepatocellular carcinoma
FA
Fatty acid
AMPK
AMP-activated protein kinase
SIK
Salt-induced kinase
PPP
Pentose phosphate pathway
SREBP
Sterol regulatory element-binding protein
ACC
Acetyl-CoA carboxylase
FASN
Fatty acid synthase
SCD1
Stearoyl-CoA desaturase 1
TG
Triglyceride
FAO
Fatty acid oxidation
LDs
Lipid droplets
Insig
Insulin-induced gene
ER
Endoplasmic reticulum
GLP-1
Glucagon-like peptide-1
LRG
Liraglutide
mTORC1
Mechanistic target of rapamycin complex 1
KO
Knockout
ACLY
ATP citrate lyase
HDACs
Histone deacetylases
CRTC
CAMP-regulated transcriptional coactivator
CREB
CAMP response element binding protein
NADPH
Nicotinamide adenine dinucleotide phosphate
Ru-5-P
Ribulose-5-phosphate
G6PD
Glucose-6-phosphate dehydrogenase
ABC
ATP binding cassette
GSEA
Gene set enrichment analysis
GO
Gene Ontology
CPT1A
Carnitine palmitoyltransferase 1A
PPAR
Peroxisome proliferator-activated receptor
SRPK2
Serine/arginine-rich protein kinase 2
ATGL
Adipose triglyceride lipase
NSCLC
Non-small cell lung cancer
SIRT1
Sirtuin 1
NAFLD
Non-alcoholic fatty liver disease
PKA
CAMP-dependent protein kinase A
ERK
Extracellular signal-regulated kinase
HMGCR
3-Hydroxy-3-methylglutaryl-CoA reductase
HSF1
Heat shock factor 1
PGE2
Prostaglandin E2

Introduction

Liver Kinase B1 (LKB1), also known as the serine-threonine kinase (STK) 11, is a tumor suppressor gene. LKB1 functions as a protein kinase which plays a vital role in maintaining energy homeostasis by phosphorylating and activating AMP-activated protein kinase (AMPK) family members [1]. LKB1 mutation is associated with the occurrence, development, and poor prognosis of several types of cancer, such as lung cancer, melanoma, cervical cancer, and hepatocellular carcinoma (HCC) [25].
Proliferation and metastasis of tumor cells involved in alteration of energy metabolism [6]. Although glucose and glutamine are important sources to maintain energy requirements for tumor cells [7], fatty acid oxidation (FAO) also provides potent metabolism support and possesses higher energy conversion efficiency [8]. In addition, excessive accumulation of lipid, which is stored in the form of lipid droplets, maintains the survival of tumor cells in energy-restricted condition [9]. LKB1 participates in the regulation of lipid synthesis, oxidation, and uptake in two ways. On the one hand, LKB1 activates downstream kinases, such as salt-induced kinase (SIK) and AMPK, which involve the activation and expression of several proteins related to lipid metabolism [10]. For instance, multiple key enzymes and transcription factors locate downstream of LKB1 and participate in de novo FA synthesis, such as sterol regulatory element-binding protein (SREBP), acetyl-CoA carboxylase (ACC), fatty acid synthase (FASN), and stearoyl-CoA desaturase-1 (SCD1) [11]. Upregulation of these FA synthesis enzymes was displayed promoting the development of several types of human cancer [12]. On the other hand, LKB1 regulates glucose metabolism via direct or indirect manners, such as the pentose phosphate pathway (PPP), which provides NADPH to lipid synthesis [13, 14]. Therefore, we focused on pathways under the regulation of LKB1 which involve lipid metabolism, and analyzed their role in the proliferation or metastasis of tumor cells. We are certain that the development and application of targeted inhibitors that process function to reverse abnormal lipid metabolism will contribute to improving prognosis in LKB1-deficient tumors.

Lipid metabolism pathways under the regulation of LKB1

LKB1-AMPK pathway

AMPK is an uppermost downstream kinase of LKB1, which functions as a central regulator of energy metabolism [15]. AMPK plays a vital role in energy metabolism and maintaining physiological functions in multiple types of cells and tissues (Fig. 1). For instance, AMPK mediates LKB1 deletion-induced enhancement of lipid-related gene expression in proliferative myoblasts [16]. Activation of AMPK significantly reduces hepatic triglyceride (TG) levels and stimulates the utilization of FA through restoring FAO [17] and inhibiting the de novo synthesis of lipid and cholesterol [18]. Activation of the LKB1-AMPK pathway also facilitates hepatocyte autophagy, which reduces lipotoxicity induced by lipid accumulation by blocking the formation of lipid droplets (LDs) [19]. Moreover, the AMPK-activated insulin-induced gene (Insig) is a protein that is bound with oxysterol in the endoplasmic reticulum (ER), which suppresses FA synthesis through inhibiting SREBP1c cleavage [20]. Liraglutide (LRG) is an agonist of Glucagon-like peptide-1 (GLP-1) receptor, which inhibits lipid synthesis by activating the AMPK-mechanistic target of rapamycin complex 1 (mTORC1)-SREBP1 pathway to ameliorate hepatic lipid accumulation [21].
AMPK negatively regulates ACC which mediates adipogenesis by catalyzing the formation of malonyl-CoA [22]. AMPK activation reduces the expression of FASN and ATP-citrate lyase (ACLY) and inhibits the activity of SREBP1, which suppresses tumor development in breast and prostate cancer [23, 24]. Together, AMPK regulates lipid metabolism by inhibiting lipid synthesis and promoting FAO, and AMPK deficiency-induced excessive lipid accumulation provides sufficient energy for the proliferation and metastasis of tumor cells. Notably, AMPK is also regulated by the ratio of AMP/ATP and calcium/calmodulin-dependent protein kinase levels [25, 26] Although LKB1 is responsible for regulating AMPK activation as the main upstream kinase, other signal pathways need to be considered when studying LKB1-AMPK pathway.

LKB1-SIK pathway

SIKs are members of the AMPK superfamily, which functions through controlling phosphorylation and the subcellular localization of class IIa histone deacetylases (IIa HDACs) and cAMP-regulated transcriptional coactivators (CRTCs) [10]. Activation of the LKB1-SIK pathway suppresses tumor development, which had been confirmed in a mouse model of lung adenocarcinoma [27]. There were 38% upregulated genes which can be attributed to SIK1/3 inactivation in all upregulated genes induced by LKB1 deficiency in non-small cell lung cancer (NSCLC) [27]. Besides, LKB1-SIK3-HDAC4 signaling functions to repair the lipid solubility defect induced by LKB1 mutants in drosophila, which is a SIK3-dependent process that isn’t affected by activation of AMPK [28]. Further study suggested that LKB1-SIK3-HDAC4 had been regarded as a potential therapeutic target for acute myeloid leukemia [29].
LKB1-SIK2 regulates the phosphorylation of CRTC2, CRTC3, and HDAC4 in adipocytes [30]. Deletion of CRTC2 reduces the expression of SREBP1c, FASN, and ACC [30]. SIK2 not only determines the properties of white adipose tissue by controlling the activity of cAMP-response element-binding protein (CREB)-CRTC2-dependent transcription [31], but also promotes cholesterol synthesis by upregulating the expression of SREBP2 in ovarian cancer cells [32]. We consider that LKB1-SIK-CRTC/HDAC4 may act as a potential target for the inhibition of tumor growth.

PPP affects lipid synthesis by providing NADPH

PPP produces ribulose-5-phosphate (Ru-5-P) and nicotinamide adenine dinucleotide phosphate (NADPH) [13]. Glucose-6-phosphate dehydrogenase (G6PD) is a rate-limiting enzyme of PPP, which is an important target of LKB1 [14]. Thus, LKB1 deficiency-induced upregulation of lipid synthesis can be attributed to intensifying PPP which provides material and facilitate the expression of genes related to FA synthesis.
PPP flux mediated by LKB1-AMPK-HDAC10-G6DP signaling had been shown to promote the growth of lung cancer cells [14]. However, LKB1-AMPK signaling is activated in breast cancer which reduces the level of G6PD transcription [33]. We speculated that upstream stimulators and tumor backgrounds underlie the different regulatory effects of LKB1 on G6PD. In the presence of glycolysis disorder, the levels of NADPH in LKB1-mutant A549 cells are higher compared to H1334 cells with normal LKB1 [34]. Nonetheless, Ru-5-P level was not changed in A549 cells with LKB1 overexpression or H1299 cell with LKB1 knocked down [14], which provides evidence that LKB1 regulates NADPH level through G6PD but Ru-5-P might be interfered with by other pathways. In addition, activation of the AMPK-SIRT1-p53 pathway increases the phosphorylation of ACC in A549 and NCI-H1299 cells [35], while downregulation of p53 increases G6PD activity in colon cancer [36], which leads us to establish a relationship between LKB1-AMPK signaling and p53-mediated G6PD regulation. Thus, further studies on the connection between LKB1, PPP, and tumor lipid metabolism are needed.

CD36 promotes lipid uptake and storage

CD36 is an important FA transporter located on the surface of various types of cells. LKB1 deficiency results in increased expression of CD36 and decreased expression of ATP binding cassette (ABC) transporter, which leads to dysfunction of cholesterol uptake, de novo synthesis, and outflow [37]. In lapatinib-resistant breast cancer cells, a gene set enrichment analysis (GSEA) and Gene Ontology (GO) enrichment analysis revealed a significant enrichment of lipid-related pathways [38]. However, upregulated of CD36 was principal, and there were no significant differences in FASN and carnitine palmitoyltransferase-1A (CPT1A) levels [38], which provides a hypothesis that CD36 mediated lipid uptake and storage, but not lipid synthesis of FAO, plays an irreplaceable role in metastatic of breast cancer cells. Deletion or inhibition of CD36 limits the uptake of FAs from the tumor microenvironment and reduces adipocyte-mediated invasion and migration of prostate cancer cells [39]. In addition, CD36 is closely related to the survival and metastasis of numerous tumors, such as lung cancer, bladder cancer, breast cancer, and melanoma [40]. Therefore, improvement of disorder lipid metabolism by blocking CD36 is an effective therapeutic strategy for inhibiting tumor development, which deserves to be expected in clinical transformation.

Other downstream lipid regulators of LKB1

Although regulation of lipid metabolism by LKB1 is a complex process (Fig. 2), energy storage is the ultimate purpose which is provided to sustain survival under energy stress conditions. Thus, LKB1 deficiency-induced abnormal lipid metabolism is displayed as enhancement in FA uptake and FA synthesis, and suppression in FAO. The LKB1-mTOR signaling inhibits differentiation of adipocyte and TG accumulation in brown adipose tissue by activating FAO and downregulating peroxisome proliferator-activated receptor γ (PPARγ) [41, 42]. Serine/arginine-rich protein kinase 2 (SRPK2) is required for de novo lipogenesis, which is under the regulation of mTOR [43]. Acetyl-CoA mediates a negative regulatory loop in the LKB1-AMPK-ACC pathway, which is a substrate of FA synthesis but participates in LKB1 acetylation [44]. We summarized that LKB1 mediated regulation of lipid metabolism processes complexity, further experimental investigations need to be conducted in different types of cancers because of the varied genetic background.

Upstream regulated molecules of the LKB1 pathway

SIRT

Sirtuin 1 (SIRT1) is required for AMPK activity. Bouchardatine is an effective inhibitor of adipogenesis, which reprograms lipid metabolism by activating SIRT1-AMPKα-ACC in rectal cancer [45], further study revealed that LKB1 mediates this signal transduction process [46, 47]. Notably, SIRT1 promotes adipocyte lipolysis in mesenteric adipose tissue but has the opposite inhibitory effect in epididymal adipose tissue [48], which illustrates certain restricted conditions, such as tissue types or genetic background, affect the role of SIRT1 in the regulation of lipid metabolism. Even so, we considered that the targeted SIRT1-LKB1 pathway remains a potential therapeutic strategy for tumors with explicit genetic background.

cAMP/PKA

cAMP-dependent protein kinase A (PKA) functions transcriptional regulation of genes related to adipogenesis. The melanocortin system plays a key role in controlling appetite and adipogenesis [49]. An investigation showed a pathway that α-Melanocyte-stimulating hormone (α-MSH)-cAMP-PKA-extracellular signal-regulated kinase (ERK) -LKB1-AMPK in hypothalamus cells, which provides a therapeutic target for lipid metabolic diseases [50]. In addition, cAMP also activates the LKB1-AMPK-SREBP pathway, which suggested that increased cAMP regulates lipid metabolism by reducing SREBPs expression [51].
PKA-LKB1 signaling pathway inhibits 3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR), ACC, and SREBP-1 through activating AMPK in insulin-resistant HepG2 cells [52]. Although LKB1 has been considered a tumor suppressor traditionally, it functions as the facilitation of tumorigenesis in thyroid cancer as the downstream mediator of PKA [53]. On the one hand, PKA opposes the negative regulation of lipolysis through phosphorylating AMPKα1 at ser173 which inhibits the activation of AMPK mediated by LKB1. On the other hand, activation of PKA directly stimulates adipocyte lipolysis, which triggers a negative feedback mechanism of AMPK. The appropriate utilization of the negative feedback loop balance between PKA and AMPK contributes to the precise regulation of lipolysis response [54]. The complex regulatory mechanism between PKA, LKB1, and AMPK provides support for maintaining metabolic balance, and may also promote tumor development, which needs to further study.

Hormones and inflammation

Hormones and inflammation are important factors that affect the development of cancers. Given that aromatase mediates hormone synthesis and several inflammatory mediators are classed as lipid, abnormal lipid metabolism also participates in the development of tumors through regulating hormones and inflammation. Prostaglandin E-2 (PGE2), an inflammatory mediator, inhibits the activity of the LKB1-AMPK pathway and regulates aromatase in breast adipose stromal cells [55]. In addition, it has been indicated that leptin induces aromatase expression by inhibiting the LKB1-AMPK pathway and subsequently nuclear translocation of CRTC2, while adiponectin has the opposite effect to leptin [56]. Vitamin D has been found to inhibit aromatase expression and local estrogen synthesis in tumor cells and adipose tissues through increasing LKB1 activity in response to the adverse effect of obesity that contributes to breast cancer growth [57]. In conclusion, the targeted LKB1 pathway may provide effective intervention for the high-risk obese people who need to prevent the occurrence and development of breast cancer more than others.

LKB1 deficiency-induced abnormal metabolism facilitates tumor progress

ACC-driven lipogenesis supports cell survival and metastasis of tumors

ACC is a major downstream lipid regulator of LKB1-AMPK signaling. ACC accelerates FAO in two CPT1-dependent manners. On the one hand, phosphorylation of ACC inactivates the ability to mediate malonyl-coA production, which promotes FAO with increased CPT1A content [58]. On the other hand, the formation of a complex between ACCα and CPT1A attenuates in the case of glucose deficiency, which boosts FAO by facilitating the transport of FA [59]. Upregulation of ACCα and p-ACC are predictors of recurrence and poor survival of tumors [59, 60]. Thus, we considered that ACC has the potential to act as a detection index, a tumor prognostic marker, or a therapeutic target.

SREBP1 maintains lipid synthesis and tumor cell viability

SREBP locates downstream of LKB1, and AMPK or SIK mediates the regulation of LKB1 for SREBPs. Activation of SREBPs regulates the expression of genes related to the key enzymes in cholesterol synthesis and FA synthesis [12]. Multiple types of tumors display activation of the mTORC1-SREBP pathway, which facilitates lipid synthesis [61]. In addition, SREBP1 is essential for the viability of cancer cells under hypoxic conditions [62]. SREBP1 is associated with the degree of malignancy and prognosis of tumors, which has been identified in metastatic melanoma [63]. Inhibition of SREBP1 increases the sensitivity of tumor therapy [64, 65], which inspires us to further develop selective SREBP1 inhibitors to suppress the development of tumors.

SCD promotes tumor growth and proliferation

SCD supports lipogenesis and desaturation of tumor cells under the reduced supply of exogenous lipids, which is important for cell viability and proliferation of tumors [66]. In addition, SCD expression is related to the survival time of patients, which can be used as a marker of tumor prognosis [67, 68]. The expression level of SCD1 in lung adenocarcinoma was higher than in adjacent normal tissues [67]. High expression of SCD usually indicates the presence of advanced disease in breast cancer and prostate cancer [66]. SCD1 also promotes the migration and invasion of tumor cells [67]. Thus, targeted inhibition of SCD has the potential to improve prognosis in tumors with high SCD expression.

Promoting effect of CPT1A on cancer

CPT1 is a rate-limiting enzyme of FAO, whose isozyme CPT1A deficiency inhibits the invasion and growth of radiotherapy-resistant breast cancer by reducing FAO [69]. A gene transcriptional splicing variant CPT1Av2 expressed in breast cancer cells interacts with HDAC1 molecules, which regulates the epigenetic inheritance of genes involved in tumor-associated cell death and invasion pathways [70]. In addition, the anti-tumor mechanism of mTORC involves the reduction of CPT1A expression and lipid catabolism [71]. Down-regulated CPT1A expression reduces lipid utilization by attenuating lipid catabolism, which contributes to the downregulation of cancer-related gene expression and apoptosis of the tumor cells [6971]. These results revealed that targeting FAO contributes to the regulation of tumor energy metabolism, and CPT1A is a potential metabolic target in cancer therapy.

ATGL affects tumor energy storage

Adipose triglyceride lipase (ATGL) is a rate-limiting enzyme in the triglyceride hydrolysis cascade, whose expression is reduced in malignant tumors [72]. The absence of ATGL induces the development of lung tumors in animal models and low levels of ATGL are associated with poor survival in patients with ovarian cancer, NSCLC, breast cancer, and stomach cancer [72]. In addition, the lipolysis activity of ATGL is crucial for the distant metastasis of tumor cells, which depends on the gradual release of stored free FA by LDs [73]. ATGL is required for the adipocyte-mediated proliferation of breast cancer cells, which uptake energy from adipocytes through the transfer of fatty acids [74]. Taken together, ATGL is considered to exert a tumor-inhibiting effect due to its deficiency in malignant tumors. However, as mechanisms of lipid metastasis provide great energy support for tumor cells, ATGL involves in tumor aggressiveness, which demonstrates further studies are required to confirm the effects of ATGL on tumor growth.

Application: targeted inhibitors of LKB1 pathways on liver diseases and tumors

Inhibitors activate the LKB1 lipid regulatory pathways

The tankyrase antagonist G007-LK was shown to play a dual role in lowering blood glucose and inhibiting the development of lung cancer by activating the LKB1-AMPK pathway [75]. A pan-Pim kinase antagonist AZD1208 with anti-cancer activity enhances LKB1 phosphorylation and reduces p-ACC, which resists adipogenesis [76]. ONC201 is an antagonist of dopamine receptor D2, which exerts anti-tumor effects in both obese and non-obese LKB1fl/fl/p53fl/fl mice with endometrial tumors. Mechanically, results from metabonomic analyses showed that ONC201 upregulated lipid synthesis [77]. Honokiol is a potential leptin antagonist, which induces SIRT1/3-mediated activation of LKB1-miR-34a signaling to antagonize tumorigenesis induced by leptin in breast cancer [78].

Activators target LKB1 downstream to release liver diseases

Several types of liver diseases relate to abnormal lipid metabolism [79]. We summarized the active factors of LKB1 and possible therapeutic effects in lipid metabolism disorders in liver diseases (Table 1). Notably, natural plant extracts and their derived compounds are beneficial to correct metabolic disorders, which applicated may exert energy stress on tumor cells to achieve suppressed effect.
Table 1
The factors acting on LKB1-mediated pathways in lipid metabolic diseases [8085]
Diseases
Factors
Mediated signalling pathways
Effects
NAFLD
Paeoniflorin α/γ-mangostin
LKB1-AMPK-SREBP1c/ FAS
Inhibiting lipid synthesis and enhancing FAO
SIRT1-LKB1-AMPK-ACC/CPT1A
ALD
Gentiopicroside
P2X7 receptor/IL1β-LKB1-AMPK-SREBP/ACC/PPARα
Reducing lipogenesis and promoting lipid oxidation
NASH
DMY
SIRT1-LKB1-AMPK-ACC
Inhibiting lipid synthesis and enhancing FAO
Liver fibrosis
HIF-1α
lncRNA-H19-LKB1-AMPK
Accelerating FAO and LDs degradation
Excessive TG
LTA
LKB1-AMPK-ACC1/SREBP1c-FAS
Enhancing oxidation and transport of lipids and reducing the synthesis of TG, cholesterol, and lipid accumulation
LKB1-AMPK-HMGCR
NAFLD Non-alcoholic fatty liver disease, ALD alcoholic liver disease, NASH non-alcoholic steatohepatitis, TG triglyceride, FAO fatty acid oxidation, LDs fat droplets, PUE Pueraria lobata, SIL Silybum marianum, DMY dihydromyricetin, HIF-1α hypoxia-inducible factor-1α, LTA L theanine

Inhibitors target LKB1 downstream to treat tumors

Several targeted inhibitors of the LKB1 pathway which relate to lipid regulation have been conducted in extensive preclinical trials in tumors. ND646, an ACC allosteric inhibitor, exerted a strong inhibitory effect on NSCLC [86]. Selective SREBP1 inhibitors reduce the expression of FASN, HMGCR, and SCD1, which decrease lipid synthesis in pancreatic cancer cells [87]. Sorafenib targets SCD1 through the ATP-AMPK-mTOR-SREBP1 signaling pathway and induces the death of HCC cells [88]. CPT1 inhibitor ST1326 counteracts the proliferation of chronic lymphocytic leukemia cells by blocking FAO and depleting acetyl-CoA in the cytoplasm [89].
Drug resistance is a practical problem in tumor treatment. ACC mutants with AMPK phosphorylation site deficiency were reported to protect head and neck squamous cell carcinoma cells to escape cetuximab-induced growth inhibition [90]. The incidence of BRAF gene mutations in melanoma is as high as 40–50%, but the therapeutic effect is not optimal when targeted BRAF. Owing to SREBP1-dependent continuous lipogenesis being a key mechanism of drug resistance induced by the BRAF mutant, simultaneous inhibition of SREBP1 enhances the sensitivity of melanoma [91], which suggests that identifying the mechanism of resistance and then synergistic use of agents will better sensitize and combat tumors.

Conclusion

LKB1 demonstrates a satisfactory ability to inhibit tumor progression, whose mutation is a mark of poor prognosis of tumor. Different cell sources and genetic backgrounds affect the expression and function of LKB1. However, LKB1 deficiency-induced abnormal lipid metabolism which is mediated by the disordering of transcriptional regulatory factors was proved to be detrimental. This review focuses on the role of LKB1-mediated pathways, which affect tumor progression through disturbing lipid metabolism. Because of the drug resistance of existing inhibitors, bimolecular targeted inhibitors and chemotherapy or radiotherapy combined with targeted inhibitors improve the therapeutic effect of tumors. In-depth studies of LKB1 are contributed to combining multiple targets for detection, which improves the accuracy of tumor hierarchical classification and stratification based on LKB1 expression. We considered that the selection of appropriate target inhibitors for sensitive tumor types may improve malignant lipid metabolism, which has the potential to inhibit tumor proliferation and metastasis.

Declarations

Conflict of interest

None.

Ethical approval

The manuscript does not contain any studies with human participants or animals.
Informed consent statements are not applicable for this article.
Open AccessThis 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/​.

Publisher's Note

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

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Jenne DE, Reimann H, Nezu J, et al. Peutz-Jeghers syndrome is caused by mutations in a novel serine threonine kinase. Nat Genet. 1998;18(1):38–43.PubMedCrossRef Jenne DE, Reimann H, Nezu J, et al. Peutz-Jeghers syndrome is caused by mutations in a novel serine threonine kinase. Nat Genet. 1998;18(1):38–43.PubMedCrossRef
2.
Zurück zum Zitat Ji H, Ramsey MR, Hayes DN, et al. LKB1 modulates lung cancer differentiation and metastasis. Nature. 2007;448(7155):807–10.PubMedCrossRef Ji H, Ramsey MR, Hayes DN, et al. LKB1 modulates lung cancer differentiation and metastasis. Nature. 2007;448(7155):807–10.PubMedCrossRef
3.
Zurück zum Zitat Zheng B, Jeong JH, Asara JM, et al. Oncogenic B-RAF negatively regulates the tumor suppressor LKB1 to promote melanoma cell proliferation. Mol Cell. 2009;33(2):237–47.PubMedPubMedCentralCrossRef Zheng B, Jeong JH, Asara JM, et al. Oncogenic B-RAF negatively regulates the tumor suppressor LKB1 to promote melanoma cell proliferation. Mol Cell. 2009;33(2):237–47.PubMedPubMedCentralCrossRef
4.
Zurück zum Zitat Zeng Q, Chen J, Li Y, et al. LKB1 inhibits HPV-associated cancer progression by targeting cellular metabolism. Oncogene. 2017;36(9):1245–55.PubMedCrossRef Zeng Q, Chen J, Li Y, et al. LKB1 inhibits HPV-associated cancer progression by targeting cellular metabolism. Oncogene. 2017;36(9):1245–55.PubMedCrossRef
5.
Zurück zum Zitat Wu CC, Wu DW, Lin YY, Lin PL, Lee H. Hepatitis B virus X protein represses LKB1 expression to promote tumor progression and poor postoperative outcome in hepatocellular carcinoma. Surgery. 2018;163(5):1040–6.PubMedCrossRef Wu CC, Wu DW, Lin YY, Lin PL, Lee H. Hepatitis B virus X protein represses LKB1 expression to promote tumor progression and poor postoperative outcome in hepatocellular carcinoma. Surgery. 2018;163(5):1040–6.PubMedCrossRef
6.
Zurück zum Zitat Kroemer G, Pouyssegur J. Tumor cell metabolism: cancer’s achilles’ heel. Cancer Cell. 2008;13(6):472–82.PubMedCrossRef Kroemer G, Pouyssegur J. Tumor cell metabolism: cancer’s achilles’ heel. Cancer Cell. 2008;13(6):472–82.PubMedCrossRef
7.
Zurück zum Zitat Zhu L, Zhu X, Wu Y. Effects of glucose metabolism, lipid metabolism, and glutamine metabolism on tumor microenvironment and clinical implications. Biomolecules. 2022;12(4):580.PubMedPubMedCentralCrossRef Zhu L, Zhu X, Wu Y. Effects of glucose metabolism, lipid metabolism, and glutamine metabolism on tumor microenvironment and clinical implications. Biomolecules. 2022;12(4):580.PubMedPubMedCentralCrossRef
8.
Zurück zum Zitat Ma Y, Temkin SM, Hawkridge AM, et al. Fatty acid oxidation: an emerging facet of metabolic transformation in cancer. Cancer Lett. 2018;435:92–100.PubMedPubMedCentralCrossRef Ma Y, Temkin SM, Hawkridge AM, et al. Fatty acid oxidation: an emerging facet of metabolic transformation in cancer. Cancer Lett. 2018;435:92–100.PubMedPubMedCentralCrossRef
9.
Zurück zum Zitat Koizume S, Miyagi Y. Lipid droplets: a key cellular organelle associated with cancer cell survival under normoxia and hypoxia. Int J Mol Sci. 2016;17(9):1430.PubMedCentralCrossRef Koizume S, Miyagi Y. Lipid droplets: a key cellular organelle associated with cancer cell survival under normoxia and hypoxia. Int J Mol Sci. 2016;17(9):1430.PubMedCentralCrossRef
10.
Zurück zum Zitat Gormand A, Berggreen C, Amar L, et al. LKB1 signalling attenuates early events of adipogenesis and responds to adipogenic cues. J Mol Endocrinol. 2014;53(1):117–30.PubMedCrossRef Gormand A, Berggreen C, Amar L, et al. LKB1 signalling attenuates early events of adipogenesis and responds to adipogenic cues. J Mol Endocrinol. 2014;53(1):117–30.PubMedCrossRef
11.
Zurück zum Zitat Seo MS, Kim JH, Kim HJ, Chang KC, Park SW. Honokiol activates the LKB1-AMPK signaling pathway and attenuates the lipid accumulation in hepatocytes. Toxicol Appl Pharmacol. 2015;284(2):113–24.PubMedCrossRef Seo MS, Kim JH, Kim HJ, Chang KC, Park SW. Honokiol activates the LKB1-AMPK signaling pathway and attenuates the lipid accumulation in hepatocytes. Toxicol Appl Pharmacol. 2015;284(2):113–24.PubMedCrossRef
12.
Zurück zum Zitat Cheng C, Geng F, Cheng X, Guo D. Lipid metabolism reprogramming and its potential targets in cancer. Cancer Commun (Lond). 2018;38(1):27.CrossRef Cheng C, Geng F, Cheng X, Guo D. Lipid metabolism reprogramming and its potential targets in cancer. Cancer Commun (Lond). 2018;38(1):27.CrossRef
14.
Zurück zum Zitat Shan C, Lu Z, Li Z, et al. 4-hydroxyphenylpyruvate dioxygenase promotes lung cancer growth via pentose phosphate pathway (PPP) flux mediated by LKB1-AMPK/HDAC10/G6PD axis. Cell Death Dis. 2019;10(7):525.PubMedPubMedCentralCrossRef Shan C, Lu Z, Li Z, et al. 4-hydroxyphenylpyruvate dioxygenase promotes lung cancer growth via pentose phosphate pathway (PPP) flux mediated by LKB1-AMPK/HDAC10/G6PD axis. Cell Death Dis. 2019;10(7):525.PubMedPubMedCentralCrossRef
15.
16.
Zurück zum Zitat Shan T, Zhang P, Bi P, Kuang S. Lkb1 deletion promotes ectopic lipid accumulation in muscle progenitor cells and mature muscles. J Cell Physiol. 2015;230(5):1033–41.PubMedPubMedCentralCrossRef Shan T, Zhang P, Bi P, Kuang S. Lkb1 deletion promotes ectopic lipid accumulation in muscle progenitor cells and mature muscles. J Cell Physiol. 2015;230(5):1033–41.PubMedPubMedCentralCrossRef
17.
Zurück zum Zitat Boudaba N, Marion A, Huet C, Pierre R, Viollet B, Foretz M. AMPK re-activation suppresses hepatic steatosis but its downregulation does not promote fatty liver development. EBioMedicine. 2018;28:194–209.PubMedPubMedCentralCrossRef Boudaba N, Marion A, Huet C, Pierre R, Viollet B, Foretz M. AMPK re-activation suppresses hepatic steatosis but its downregulation does not promote fatty liver development. EBioMedicine. 2018;28:194–209.PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Esquejo RM, Salatto CT, Delmore J, et al. Activation of liver AMPK with PF-06409577 corrects NAFLD and lowers cholesterol in rodent and primate preclinical models. EBioMedicine. 2018;31:122–32.PubMedPubMedCentralCrossRef Esquejo RM, Salatto CT, Delmore J, et al. Activation of liver AMPK with PF-06409577 corrects NAFLD and lowers cholesterol in rodent and primate preclinical models. EBioMedicine. 2018;31:122–32.PubMedPubMedCentralCrossRef
19.
Zurück zum Zitat Huang Y, Liu F, Zhang F, Liu P, Xu T, Ding W. Vanadium(IV)-chlorodipicolinate alleviates hepatic lipid accumulation by inducing autophagy via the LKB1/AMPK signaling pathway in vitro and in vivo. J Inorg Biochem. 2018;183:66–76.PubMedCrossRef Huang Y, Liu F, Zhang F, Liu P, Xu T, Ding W. Vanadium(IV)-chlorodipicolinate alleviates hepatic lipid accumulation by inducing autophagy via the LKB1/AMPK signaling pathway in vitro and in vivo. J Inorg Biochem. 2018;183:66–76.PubMedCrossRef
20.
Zurück zum Zitat Han Y, Hu Z, Cui A, et al. Post-translational regulation of lipogenesis via AMPK-dependent phosphorylation of insulin-induced gene. Nat Commun. 2019;10(1):623.PubMedPubMedCentralCrossRef Han Y, Hu Z, Cui A, et al. Post-translational regulation of lipogenesis via AMPK-dependent phosphorylation of insulin-induced gene. Nat Commun. 2019;10(1):623.PubMedPubMedCentralCrossRef
21.
Zurück zum Zitat Hao T, Chen H, Wu S, Tian H. LRG ameliorates steatohepatitis by activating the AMPK/mTOR/SREBP1 signaling pathway in C57BL/6J mice fed a high-fat diet. Mol Med Rep. 2019;20(1):701–8.PubMed Hao T, Chen H, Wu S, Tian H. LRG ameliorates steatohepatitis by activating the AMPK/mTOR/SREBP1 signaling pathway in C57BL/6J mice fed a high-fat diet. Mol Med Rep. 2019;20(1):701–8.PubMed
22.
Zurück zum Zitat Thomson DM, Brown JD, Fillmore N, et al. LKB1 and the regulation of malonyl-CoA and fatty acid oxidation in muscle. Am J Physiol Endocrinol Metab. 2007;293(6):E1572–9.PubMedCrossRef Thomson DM, Brown JD, Fillmore N, et al. LKB1 and the regulation of malonyl-CoA and fatty acid oxidation in muscle. Am J Physiol Endocrinol Metab. 2007;293(6):E1572–9.PubMedCrossRef
23.
Zurück zum Zitat Zhang ZG, Zhang HS, Sun HL, Liu HY, Liu MY, Zhou Z. KDM5B promotes breast cancer cell proliferation and migration via AMPK-mediated lipid metabolism reprogramming. Exp Cell Res. 2019;379(2):182–90.PubMedCrossRef Zhang ZG, Zhang HS, Sun HL, Liu HY, Liu MY, Zhou Z. KDM5B promotes breast cancer cell proliferation and migration via AMPK-mediated lipid metabolism reprogramming. Exp Cell Res. 2019;379(2):182–90.PubMedCrossRef
24.
Zurück zum Zitat Zheng Y, Jin J, Gao Y, Luo C, Wu X, Liu J. Phospholipase Cε regulates prostate cancer lipid metabolism and proliferation by targeting AMP-activated protein kinase (AMPK)/sterol regulatory element-binding protein 1 (SREBP-1) signaling pathway. Med Sci Monit. 2020;26: e924328.PubMedPubMedCentralCrossRef Zheng Y, Jin J, Gao Y, Luo C, Wu X, Liu J. Phospholipase Cε regulates prostate cancer lipid metabolism and proliferation by targeting AMP-activated protein kinase (AMPK)/sterol regulatory element-binding protein 1 (SREBP-1) signaling pathway. Med Sci Monit. 2020;26: e924328.PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Stenesen D, Suh JM, Seo J, et al. Adenosine nucleotide biosynthesis and AMPK regulate adult life span and mediate the longevity benefit of caloric restriction in flies. Cell Metab. 2013;17(1):101–12.PubMedPubMedCentralCrossRef Stenesen D, Suh JM, Seo J, et al. Adenosine nucleotide biosynthesis and AMPK regulate adult life span and mediate the longevity benefit of caloric restriction in flies. Cell Metab. 2013;17(1):101–12.PubMedPubMedCentralCrossRef
26.
Zurück zum Zitat Wang MD, Wu H, Huang S, et al. HBx regulates fatty acid oxidation to promote hepatocellular carcinoma survival during metabolic stress. Oncotarget. 2016;7(6):6711–26.PubMedPubMedCentralCrossRef Wang MD, Wu H, Huang S, et al. HBx regulates fatty acid oxidation to promote hepatocellular carcinoma survival during metabolic stress. Oncotarget. 2016;7(6):6711–26.PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Hollstein PE, Eichner LJ, Brun SN, et al. The AMPK-related kinases SIK1 and SIK3 mediate key tumor-suppressive effects of LKB1 in NSCLC. Cancer Discov. 2019;9(11):1606–27.PubMedPubMedCentralCrossRef Hollstein PE, Eichner LJ, Brun SN, et al. The AMPK-related kinases SIK1 and SIK3 mediate key tumor-suppressive effects of LKB1 in NSCLC. Cancer Discov. 2019;9(11):1606–27.PubMedPubMedCentralCrossRef
28.
Zurück zum Zitat Choi S, Lim DS, Chung J. Feeding and fasting signals converge on the LKB1-SIK3 pathway to regulate lipid metabolism in drosophila. PLoS Genet. 2015;11(5): e1005263.PubMedPubMedCentralCrossRef Choi S, Lim DS, Chung J. Feeding and fasting signals converge on the LKB1-SIK3 pathway to regulate lipid metabolism in drosophila. PLoS Genet. 2015;11(5): e1005263.PubMedPubMedCentralCrossRef
29.
Zurück zum Zitat Tarumoto Y, Lu B, Somerville TDD, et al. LKB1, salt-inducible kinases, and MEF2C are linked dependencies in acute myeloid leukemia. Mol Cell. 2018;69(6):1017–27.PubMedPubMedCentralCrossRef Tarumoto Y, Lu B, Somerville TDD, et al. LKB1, salt-inducible kinases, and MEF2C are linked dependencies in acute myeloid leukemia. Mol Cell. 2018;69(6):1017–27.PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Henriksson E, Säll J, Gormand A, et al. SIK2 regulates CRTCs, HDAC4 and glucose uptake in adipocytes. J Cell Sci. 2015;128(3):472–86.PubMedPubMedCentral Henriksson E, Säll J, Gormand A, et al. SIK2 regulates CRTCs, HDAC4 and glucose uptake in adipocytes. J Cell Sci. 2015;128(3):472–86.PubMedPubMedCentral
31.
Zurück zum Zitat Park J, Yoon YS, Han HS, et al. SIK2 is critical in the regulation of lipid homeostasis and adipogenesis in vivo. Diabetes. 2014;63(11):3659–73.PubMedCrossRef Park J, Yoon YS, Han HS, et al. SIK2 is critical in the regulation of lipid homeostasis and adipogenesis in vivo. Diabetes. 2014;63(11):3659–73.PubMedCrossRef
32.
Zurück zum Zitat Zhao J, Zhang X, Gao T, et al. SIK2 enhances synthesis of fatty acid and cholesterol in ovarian cancer cells and tumor growth through PI3K/Akt signaling pathway. Cell Death Dis. 2020;11(1):25.PubMedPubMedCentralCrossRef Zhao J, Zhang X, Gao T, et al. SIK2 enhances synthesis of fatty acid and cholesterol in ovarian cancer cells and tumor growth through PI3K/Akt signaling pathway. Cell Death Dis. 2020;11(1):25.PubMedPubMedCentralCrossRef
33.
Zurück zum Zitat Yang L, He Z, Yao J, et al. Regulation of AMPK-related glycolipid metabolism imbalances redox homeostasis and inhibits anchorage independent growth in human breast cancer cells [published correction appears in Redox Biol. 2020 Jan; 28:101382]. Redox Biol. 2018;17:180–91.PubMedPubMedCentralCrossRef Yang L, He Z, Yao J, et al. Regulation of AMPK-related glycolipid metabolism imbalances redox homeostasis and inhibits anchorage independent growth in human breast cancer cells [published correction appears in Redox Biol. 2020 Jan; 28:101382]. Redox Biol. 2018;17:180–91.PubMedPubMedCentralCrossRef
34.
35.
Zurück zum Zitat Zhang Y, Sun C, Xiao G, et al. S-nitrosylation of the peroxiredoxin-2 promotes S-nitrosoglutathione-mediated lung cancer cells apoptosis via AMPK-SIRT1 pathway. Cell Death Dis. 2019;10(5):329.PubMedPubMedCentralCrossRef Zhang Y, Sun C, Xiao G, et al. S-nitrosylation of the peroxiredoxin-2 promotes S-nitrosoglutathione-mediated lung cancer cells apoptosis via AMPK-SIRT1 pathway. Cell Death Dis. 2019;10(5):329.PubMedPubMedCentralCrossRef
36.
Zurück zum Zitat Zhang X, Zhang X, Li Y, et al. PAK4 regulates G6PD activity by p53 degradation involving colon cancer cell growth. Cell Death Dis. 2017;8(5): e2820.PubMedPubMedCentralCrossRef Zhang X, Zhang X, Li Y, et al. PAK4 regulates G6PD activity by p53 degradation involving colon cancer cell growth. Cell Death Dis. 2017;8(5): e2820.PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Timilshina M, You Z, Lacher SM, et al. Activation of mevalonate pathway via LKB1 is essential for stability of treg cells. Cell Rep. 2019;27(10):2948-2961.e7.PubMedCrossRef Timilshina M, You Z, Lacher SM, et al. Activation of mevalonate pathway via LKB1 is essential for stability of treg cells. Cell Rep. 2019;27(10):2948-2961.e7.PubMedCrossRef
38.
Zurück zum Zitat Feng WW, Wilkins O, Bang S, et al. CD36-mediated metabolic rewiring of breast cancer cells promotes resistance to HER2-targeted therapies. Cell Rep. 2019;29(11):3405–20.PubMedPubMedCentralCrossRef Feng WW, Wilkins O, Bang S, et al. CD36-mediated metabolic rewiring of breast cancer cells promotes resistance to HER2-targeted therapies. Cell Rep. 2019;29(11):3405–20.PubMedPubMedCentralCrossRef
39.
Zurück zum Zitat Watt MJ, Clark AK, Selth LA, et al. Suppressing fatty acid uptake has therapeutic effects in preclinical models of prostate cancer. Sci Transl Med. 2019;11(478):5758.CrossRef Watt MJ, Clark AK, Selth LA, et al. Suppressing fatty acid uptake has therapeutic effects in preclinical models of prostate cancer. Sci Transl Med. 2019;11(478):5758.CrossRef
40.
Zurück zum Zitat Pascual G, Avgustinova A, Mejetta S, et al. Targeting metastasis-initiating cells through the fatty acid receptor CD36. Nature. 2017;541(7635):41–5.PubMedCrossRef Pascual G, Avgustinova A, Mejetta S, et al. Targeting metastasis-initiating cells through the fatty acid receptor CD36. Nature. 2017;541(7635):41–5.PubMedCrossRef
41.
Zurück zum Zitat Shan T, Zhang P, Jiang Q, Xiong Y, Wang Y, Kuang S. Adipocyte-specific deletion of mTOR inhibits adipose tissue development and causes insulin resistance in mice. Diabetologia. 2016;59(9):1995–2004.PubMedPubMedCentralCrossRef Shan T, Zhang P, Jiang Q, Xiong Y, Wang Y, Kuang S. Adipocyte-specific deletion of mTOR inhibits adipose tissue development and causes insulin resistance in mice. Diabetologia. 2016;59(9):1995–2004.PubMedPubMedCentralCrossRef
42.
Zurück zum Zitat Xiong Y, Xu Z, Wang Y, Kuang S, Shan T. Adipocyte-specific DKO of Lkb1 and mTOR protects mice against HFD-induced obesity, but results in insulin resistance. J Lipid Res. 2018;59(6):974–81.PubMedPubMedCentralCrossRef Xiong Y, Xu Z, Wang Y, Kuang S, Shan T. Adipocyte-specific DKO of Lkb1 and mTOR protects mice against HFD-induced obesity, but results in insulin resistance. J Lipid Res. 2018;59(6):974–81.PubMedPubMedCentralCrossRef
43.
Zurück zum Zitat Lee G, Zheng Y, Cho S, et al. Post-transcriptional regulation of de novo lipogenesis by mTORC1-S6K1-SRPK2 signaling. Cell. 2017;171(7):1545–58.PubMedPubMedCentralCrossRef Lee G, Zheng Y, Cho S, et al. Post-transcriptional regulation of de novo lipogenesis by mTORC1-S6K1-SRPK2 signaling. Cell. 2017;171(7):1545–58.PubMedPubMedCentralCrossRef
44.
Zurück zum Zitat Vancura A, Nagar S, Kaur P, Bu P, Bhagwat M, Vancurova I. Reciprocal regulation of AMPK/SNF1 and protein acetylation. Int J Mol Sci. 2018;19(11):3314.PubMedCentralCrossRef Vancura A, Nagar S, Kaur P, Bu P, Bhagwat M, Vancurova I. Reciprocal regulation of AMPK/SNF1 and protein acetylation. Int J Mol Sci. 2018;19(11):3314.PubMedCentralCrossRef
45.
Zurück zum Zitat Xu YH, Song QQ, Li C, et al. Bouchardatine suppresses rectal cancer in mice by disrupting its metabolic pathways via activating the SIRT1-PGC-1α-UCP2 axis. Eur J Pharmacol. 2019;854:328–37.PubMedCrossRef Xu YH, Song QQ, Li C, et al. Bouchardatine suppresses rectal cancer in mice by disrupting its metabolic pathways via activating the SIRT1-PGC-1α-UCP2 axis. Eur J Pharmacol. 2019;854:328–37.PubMedCrossRef
46.
Zurück zum Zitat Park EJ, Kim YM, Kim HJ, et al. (S)YS-51, a novel isoquinoline alkaloid, attenuates obesity-associated non-alcoholic fatty liver disease in mice by suppressing lipogenesis, inflammation and coagulation. Eur J Pharmacol. 2016;788:200–9.PubMedCrossRef Park EJ, Kim YM, Kim HJ, et al. (S)YS-51, a novel isoquinoline alkaloid, attenuates obesity-associated non-alcoholic fatty liver disease in mice by suppressing lipogenesis, inflammation and coagulation. Eur J Pharmacol. 2016;788:200–9.PubMedCrossRef
47.
Zurück zum Zitat Long JK, Dai W, Zheng YW, Zhao SP. miR-122 promotes hepatic lipogenesis via inhibiting the LKB1/AMPK pathway by targeting Sirt1 in non-alcoholic fatty liver disease. Mol Med. 2019;25(1):26.PubMedPubMedCentralCrossRef Long JK, Dai W, Zheng YW, Zhao SP. miR-122 promotes hepatic lipogenesis via inhibiting the LKB1/AMPK pathway by targeting Sirt1 in non-alcoholic fatty liver disease. Mol Med. 2019;25(1):26.PubMedPubMedCentralCrossRef
48.
Zurück zum Zitat Cheng J, Liu C, Hu K, et al. Ablation of systemic SIRT1 activity promotes nonalcoholic fatty liver disease by affecting liver-mesenteric adipose tissue fatty acid mobilization. Biochim Biophys Acta Mol Basis Dis. 2017;1863(11):2783–90.PubMedCrossRef Cheng J, Liu C, Hu K, et al. Ablation of systemic SIRT1 activity promotes nonalcoholic fatty liver disease by affecting liver-mesenteric adipose tissue fatty acid mobilization. Biochim Biophys Acta Mol Basis Dis. 2017;1863(11):2783–90.PubMedCrossRef
49.
Zurück zum Zitat MacNeil DJ, Howard AD, Guan X, et al. The role of melanocortins in body weight regulation: opportunities for the treatment of obesity. Eur J Pharmacol. 2002;450(1):93–109.PubMedCrossRef MacNeil DJ, Howard AD, Guan X, et al. The role of melanocortins in body weight regulation: opportunities for the treatment of obesity. Eur J Pharmacol. 2002;450(1):93–109.PubMedCrossRef
50.
Zurück zum Zitat Damm E, Buech TR, Gudermann T, Breit A. Melanocortin-induced PKA activation inhibits AMPK activity via ERK-1/2 and LKB-1 in hypothalamic GT1-7 cells. Mol Endocrinol. 2012;26(4):643–54.PubMedPubMedCentralCrossRef Damm E, Buech TR, Gudermann T, Breit A. Melanocortin-induced PKA activation inhibits AMPK activity via ERK-1/2 and LKB-1 in hypothalamic GT1-7 cells. Mol Endocrinol. 2012;26(4):643–54.PubMedPubMedCentralCrossRef
51.
Zurück zum Zitat Lu J, Fang B, Huang Y, Tao S, Sun B, Guan S, Jin Y. Epigallocatechin-3-gallate protects against 1,3-dichloro-2-propanol-induced lipid accumulation in C57BL/6J mice. Life Sci. 2018;209:324–31.PubMedCrossRef Lu J, Fang B, Huang Y, Tao S, Sun B, Guan S, Jin Y. Epigallocatechin-3-gallate protects against 1,3-dichloro-2-propanol-induced lipid accumulation in C57BL/6J mice. Life Sci. 2018;209:324–31.PubMedCrossRef
52.
Zurück zum Zitat Wang X, Jiang H, Zhang N, Cai C, Li G, Hao J, Yu G. Anti-diabetic activities of agaropectin-derived oligosaccharides from Gloiopeltis furcata via regulation of mitochondrial function. Carbohydr Polym. 2020;229: 115482.PubMedCrossRef Wang X, Jiang H, Zhang N, Cai C, Li G, Hao J, Yu G. Anti-diabetic activities of agaropectin-derived oligosaccharides from Gloiopeltis furcata via regulation of mitochondrial function. Carbohydr Polym. 2020;229: 115482.PubMedCrossRef
53.
Zurück zum Zitat Kari S, Vasko VV, Priya S, Kirschner LS. PKA activates AMPK through LKB1 signaling in follicular thyroid cancer. Front Endocrinol (Lausanne). 2019;10:769.CrossRef Kari S, Vasko VV, Priya S, Kirschner LS. PKA activates AMPK through LKB1 signaling in follicular thyroid cancer. Front Endocrinol (Lausanne). 2019;10:769.CrossRef
54.
Zurück zum Zitat Djouder N, Tuerk RD, Suter M, et al. PKA phosphorylates and inactivates AMPKalpha to promote efficient lipolysis. EMBO J. 2010;29(2):469–81.PubMedCrossRef Djouder N, Tuerk RD, Suter M, et al. PKA phosphorylates and inactivates AMPKalpha to promote efficient lipolysis. EMBO J. 2010;29(2):469–81.PubMedCrossRef
55.
Zurück zum Zitat Zahid H, Simpson ER, Brown KA. Inflammation, dysregulated metabolism and aromatase in obesity and breast cancer. Curr Opin Pharmacol. 2016;31:90–6.PubMedCrossRef Zahid H, Simpson ER, Brown KA. Inflammation, dysregulated metabolism and aromatase in obesity and breast cancer. Curr Opin Pharmacol. 2016;31:90–6.PubMedCrossRef
56.
Zurück zum Zitat Brown KA, McInnes KJ, Hunger NI, Oakhill JS, Steinberg GR, Simpson ER. Subcellular localization of cyclic AMP-responsive element binding protein-regulated transcription coactivator 2 provides a link between obesity and breast cancer in postmenopausal women. Cancer Res. 2009;69(13):5392–9.PubMedCrossRef Brown KA, McInnes KJ, Hunger NI, Oakhill JS, Steinberg GR, Simpson ER. Subcellular localization of cyclic AMP-responsive element binding protein-regulated transcription coactivator 2 provides a link between obesity and breast cancer in postmenopausal women. Cancer Res. 2009;69(13):5392–9.PubMedCrossRef
57.
Zurück zum Zitat Swami S, Krishnan AV, Williams J, et al. Vitamin D mitigates the adverse effects of obesity on breast cancer in mice. Endocr Relat Cancer. 2016;23(4):251–64.PubMedPubMedCentralCrossRef Swami S, Krishnan AV, Williams J, et al. Vitamin D mitigates the adverse effects of obesity on breast cancer in mice. Endocr Relat Cancer. 2016;23(4):251–64.PubMedPubMedCentralCrossRef
58.
Zurück zum Zitat Yao Q, Li S, Cheng X, Zou Y, Shen Y, Zhang S. Yin Zhi Huang, a traditional Chinese herbal formula, ameliorates diet-induced obesity and hepatic steatosis by activating the AMPK/SREBP-1 and the AMPK/ACC/CPT1A pathways. Ann Transl Med. 2020;8(5):231.PubMedPubMedCentralCrossRef Yao Q, Li S, Cheng X, Zou Y, Shen Y, Zhang S. Yin Zhi Huang, a traditional Chinese herbal formula, ameliorates diet-induced obesity and hepatic steatosis by activating the AMPK/SREBP-1 and the AMPK/ACC/CPT1A pathways. Ann Transl Med. 2020;8(5):231.PubMedPubMedCentralCrossRef
59.
Zurück zum Zitat Wang MD, Wu H, Fu GB, et al. Acetyl-coenzyme A carboxylase alpha promotion of glucose-mediated fatty acid synthesis enhances survival of hepatocellular carcinoma in mice and patients. Hepatology. 2016;63(4):1272–86.PubMedCrossRef Wang MD, Wu H, Fu GB, et al. Acetyl-coenzyme A carboxylase alpha promotion of glucose-mediated fatty acid synthesis enhances survival of hepatocellular carcinoma in mice and patients. Hepatology. 2016;63(4):1272–86.PubMedCrossRef
60.
Zurück zum Zitat Indraccolo S, De Salvo GL, Verza M, et al. Phosphorylated acetyl-CoA carboxylase is associated with clinical benefit with regorafenib in relapsed glioblastoma: REGOMA trial biomarker analysis. Clin Cancer Res. 2020;26(17):4478–84.PubMedCrossRef Indraccolo S, De Salvo GL, Verza M, et al. Phosphorylated acetyl-CoA carboxylase is associated with clinical benefit with regorafenib in relapsed glioblastoma: REGOMA trial biomarker analysis. Clin Cancer Res. 2020;26(17):4478–84.PubMedCrossRef
61.
Zurück zum Zitat Ricoult SJ, Yecies JL, Ben-Sahra I, Manning BD. Oncogenic PI3K and K-Ras stimulate de novo lipid synthesis through mTORC1 and SREBP. Oncogene. 2016;35(10):1250–60.PubMedCrossRef Ricoult SJ, Yecies JL, Ben-Sahra I, Manning BD. Oncogenic PI3K and K-Ras stimulate de novo lipid synthesis through mTORC1 and SREBP. Oncogene. 2016;35(10):1250–60.PubMedCrossRef
62.
Zurück zum Zitat Lewis CA, Brault C, Peck B, et al. SREBP maintains lipid biosynthesis and viability of cancer cells under lipid- and oxygen-deprived conditions and defines a gene signature associated with poor survival in glioblastoma multiforme. Oncogene. 2015;34(40):5128–40.PubMedCrossRef Lewis CA, Brault C, Peck B, et al. SREBP maintains lipid biosynthesis and viability of cancer cells under lipid- and oxygen-deprived conditions and defines a gene signature associated with poor survival in glioblastoma multiforme. Oncogene. 2015;34(40):5128–40.PubMedCrossRef
63.
Zurück zum Zitat Giampietri C, Petrungaro S, Cordella M, et al. Lipid Storage and autophagy in melanoma cancer cells. Int J Mol Sci. 2017;18(6):1271.PubMedCentralCrossRef Giampietri C, Petrungaro S, Cordella M, et al. Lipid Storage and autophagy in melanoma cancer cells. Int J Mol Sci. 2017;18(6):1271.PubMedCentralCrossRef
64.
Zurück zum Zitat Shi Y, Fan Y, Hu Y, et al. α-Mangostin suppresses the de novo lipogenesis and enhances the chemotherapeutic response to gemcitabine in gallbladder carcinoma cells via targeting the AMPK/SREBP1 cascades. J Cell Mol Med. 2020;24(1):760–71.PubMedCrossRef Shi Y, Fan Y, Hu Y, et al. α-Mangostin suppresses the de novo lipogenesis and enhances the chemotherapeutic response to gemcitabine in gallbladder carcinoma cells via targeting the AMPK/SREBP1 cascades. J Cell Mol Med. 2020;24(1):760–71.PubMedCrossRef
65.
Zurück zum Zitat Ma X, Zhao T, Yan H, et al. Fatostatin reverses progesterone resistance by inhibiting the SREBP1-NF-κB pathway in endometrial carcinoma. Cell Death Dis. 2021;12(6):544.PubMedPubMedCentralCrossRef Ma X, Zhao T, Yan H, et al. Fatostatin reverses progesterone resistance by inhibiting the SREBP1-NF-κB pathway in endometrial carcinoma. Cell Death Dis. 2021;12(6):544.PubMedPubMedCentralCrossRef
66.
Zurück zum Zitat Peck B, Schug ZT, Zhang Q, et al. Inhibition of fatty acid desaturation is detrimental to cancer cell survival in metabolically compromised environments. Cancer Metab. 2016;4:6.PubMedPubMedCentralCrossRef Peck B, Schug ZT, Zhang Q, et al. Inhibition of fatty acid desaturation is detrimental to cancer cell survival in metabolically compromised environments. Cancer Metab. 2016;4:6.PubMedPubMedCentralCrossRef
67.
Zurück zum Zitat Huang J, Fan XX, He J, et al. SCD1 is associated with tumor promotion, late stage and poor survival in lung adenocarcinoma. Oncotarget. 2016;7(26):39970–9.PubMedPubMedCentralCrossRef Huang J, Fan XX, He J, et al. SCD1 is associated with tumor promotion, late stage and poor survival in lung adenocarcinoma. Oncotarget. 2016;7(26):39970–9.PubMedPubMedCentralCrossRef
68.
Zurück zum Zitat Wang J, Xu Y, Zhu L, et al. High expression of stearoyl-CoA desaturase 1 predicts poor prognosis in patients with clear-cell renal cell carcinoma. PLoS ONE. 2016;11(11): e0166231.PubMedPubMedCentralCrossRef Wang J, Xu Y, Zhu L, et al. High expression of stearoyl-CoA desaturase 1 predicts poor prognosis in patients with clear-cell renal cell carcinoma. PLoS ONE. 2016;11(11): e0166231.PubMedPubMedCentralCrossRef
69.
70.
Zurück zum Zitat Pucci S, Zonetti MJ, Fisco T, et al. Carnitine palmitoyl transferase-1A (CPT1A): a new tumor specific target in human breast cancer. Oncotarget. 2016;7(15):19982–96.PubMedPubMedCentralCrossRef Pucci S, Zonetti MJ, Fisco T, et al. Carnitine palmitoyl transferase-1A (CPT1A): a new tumor specific target in human breast cancer. Oncotarget. 2016;7(15):19982–96.PubMedPubMedCentralCrossRef
71.
Zurück zum Zitat Qian J, Chen Y, Meng T, et al. Molecular regulation of apoptotic machinery and lipid metabolism by mTORC1/mTORC2 dual inhibitors in preclinical models of HER2+/PIK3CAmut breast cancer. Oncotarget. 2016;7(41):67071–86.PubMedPubMedCentralCrossRef Qian J, Chen Y, Meng T, et al. Molecular regulation of apoptotic machinery and lipid metabolism by mTORC1/mTORC2 dual inhibitors in preclinical models of HER2+/PIK3CAmut breast cancer. Oncotarget. 2016;7(41):67071–86.PubMedPubMedCentralCrossRef
72.
Zurück zum Zitat Al-Zoughbi W, Pichler M, Gorkiewicz G, et al. Loss of adipose triglyceride lipase is associated with human cancer and induces mouse pulmonary neoplasia. Oncotarget. 2016;7(23):33832–40.PubMedPubMedCentralCrossRef Al-Zoughbi W, Pichler M, Gorkiewicz G, et al. Loss of adipose triglyceride lipase is associated with human cancer and induces mouse pulmonary neoplasia. Oncotarget. 2016;7(23):33832–40.PubMedPubMedCentralCrossRef
73.
Zurück zum Zitat Wang YY, Attané C, Milhas D, et al. Mammary adipocytes stimulate breast cancer invasion through metabolic remodeling of tumor cells. JCI Insight. 2017;2(4): e87489.PubMedPubMedCentralCrossRef Wang YY, Attané C, Milhas D, et al. Mammary adipocytes stimulate breast cancer invasion through metabolic remodeling of tumor cells. JCI Insight. 2017;2(4): e87489.PubMedPubMedCentralCrossRef
74.
Zurück zum Zitat Balaban S, Shearer RF, Lee LS, et al. Adipocyte lipolysis links obesity to breast cancer growth: adipocyte-derived fatty acids drive breast cancer cell proliferation and migration. Cancer Metab. 2017;5:1.PubMedPubMedCentralCrossRef Balaban S, Shearer RF, Lee LS, et al. Adipocyte lipolysis links obesity to breast cancer growth: adipocyte-derived fatty acids drive breast cancer cell proliferation and migration. Cancer Metab. 2017;5:1.PubMedPubMedCentralCrossRef
75.
Zurück zum Zitat Li N, Wang Y, Neri S, et al. Tankyrase disrupts metabolic homeostasis and promotes tumorigenesis by inhibiting LKB1-AMPK signalling. Nat Commun. 2019;10(1):4363.PubMedPubMedCentralCrossRef Li N, Wang Y, Neri S, et al. Tankyrase disrupts metabolic homeostasis and promotes tumorigenesis by inhibiting LKB1-AMPK signalling. Nat Commun. 2019;10(1):4363.PubMedPubMedCentralCrossRef
76.
Zurück zum Zitat Park YK, Obiang-Obounou BW, Lee KB, Choi JS, Jang BC. AZD1208, a pan-Pim kinase inhibitor, inhibits adipogenesis and induces lipolysis in 3T3-L1 adipocytes. J Cell Mol Med. 2018;22(4):2488–97.PubMedPubMedCentralCrossRef Park YK, Obiang-Obounou BW, Lee KB, Choi JS, Jang BC. AZD1208, a pan-Pim kinase inhibitor, inhibits adipogenesis and induces lipolysis in 3T3-L1 adipocytes. J Cell Mol Med. 2018;22(4):2488–97.PubMedPubMedCentralCrossRef
77.
Zurück zum Zitat Pierce SR, Fang Z, Yin Y, et al. Targeting dopamine receptor D2 as a novel therapeutic strategy in endometrial cancer. J Exp Clin Cancer Res. 2021;40(1):61.PubMedPubMedCentralCrossRef Pierce SR, Fang Z, Yin Y, et al. Targeting dopamine receptor D2 as a novel therapeutic strategy in endometrial cancer. J Exp Clin Cancer Res. 2021;40(1):61.PubMedPubMedCentralCrossRef
78.
Zurück zum Zitat Avtanski DB, Nagalingam A, Bonner MY, Arbiser JL, Saxena NK, Sharma D. Honokiol activates LKB1-miR-34a axis and antagonizes the oncogenic actions of leptin in breast cancer. Oncotarget. 2015;6(30):29947–62.PubMedPubMedCentralCrossRef Avtanski DB, Nagalingam A, Bonner MY, Arbiser JL, Saxena NK, Sharma D. Honokiol activates LKB1-miR-34a axis and antagonizes the oncogenic actions of leptin in breast cancer. Oncotarget. 2015;6(30):29947–62.PubMedPubMedCentralCrossRef
79.
Zurück zum Zitat Watanabe S, Yaginuma R, Ikejima K, Miyazaki A. Liver diseases and metabolic syndrome. J Gastroenterol. 2008;43(7):509–18.PubMedCrossRef Watanabe S, Yaginuma R, Ikejima K, Miyazaki A. Liver diseases and metabolic syndrome. J Gastroenterol. 2008;43(7):509–18.PubMedCrossRef
80.
Zurück zum Zitat Li YC, Qiao JY, Wang BY, Bai M, Shen JD, Cheng YX. Paeoniflorin ameliorates fructose-induced insulin resistance and hepatic steatosis by activating LKB1/AMPK and AKT pathways. Nutrients. 2018;10(8):1024.PubMedCentralCrossRef Li YC, Qiao JY, Wang BY, Bai M, Shen JD, Cheng YX. Paeoniflorin ameliorates fructose-induced insulin resistance and hepatic steatosis by activating LKB1/AMPK and AKT pathways. Nutrients. 2018;10(8):1024.PubMedCentralCrossRef
81.
Zurück zum Zitat Gu L, Cai N, Lyu Y, et al. γ-Mangostin ameliorates free fatty acid-induced lipid accumulation via the SIRT1/LKB1/AMPK pathway in HepG2 and L02 cells. J Agric Food Chem. 2019;67(50):13929–38.PubMedCrossRef Gu L, Cai N, Lyu Y, et al. γ-Mangostin ameliorates free fatty acid-induced lipid accumulation via the SIRT1/LKB1/AMPK pathway in HepG2 and L02 cells. J Agric Food Chem. 2019;67(50):13929–38.PubMedCrossRef
82.
Zurück zum Zitat Li X, Zhang Y, Jin Q, et al. Liver kinase B1/AMP-activated protein kinase-mediated regulation by gentiopicroside ameliorates P2X7 receptor-dependent alcoholic hepatosteatosis. Br J Pharmacol. 2018;175(9):1451–70.PubMedPubMedCentralCrossRef Li X, Zhang Y, Jin Q, et al. Liver kinase B1/AMP-activated protein kinase-mediated regulation by gentiopicroside ameliorates P2X7 receptor-dependent alcoholic hepatosteatosis. Br J Pharmacol. 2018;175(9):1451–70.PubMedPubMedCentralCrossRef
83.
Zurück zum Zitat Zeng Y, Hua YQ, Wang W, Zhang H, Xu XL. Modulation of SIRT1-mediated signaling cascades in the liver contributes to the amelioration of nonalcoholic steatohepatitis in high fat fed middle-aged LDL receptor knockout mice by dihydromyricetin. Biochem Pharmacol. 2020;175: 113927.PubMedCrossRef Zeng Y, Hua YQ, Wang W, Zhang H, Xu XL. Modulation of SIRT1-mediated signaling cascades in the liver contributes to the amelioration of nonalcoholic steatohepatitis in high fat fed middle-aged LDL receptor knockout mice by dihydromyricetin. Biochem Pharmacol. 2020;175: 113927.PubMedCrossRef
84.
Zurück zum Zitat Wang Z, Yang X, Kai J, et al. HIF-1α-upregulated lncRNA-H19 regulates lipid droplet metabolism through the AMPKα pathway in hepatic stellate cells. Life Sci. 2020;255: 117818.PubMedCrossRef Wang Z, Yang X, Kai J, et al. HIF-1α-upregulated lncRNA-H19 regulates lipid droplet metabolism through the AMPKα pathway in hepatic stellate cells. Life Sci. 2020;255: 117818.PubMedCrossRef
85.
Zurück zum Zitat Lin L, Zeng L, Liu A, et al. l-Theanine regulates glucose, lipid, and protein metabolism via insulin and AMP-activated protein kinase signaling pathways. Food Funct. 2020;11(2):1798–809.PubMedCrossRef Lin L, Zeng L, Liu A, et al. l-Theanine regulates glucose, lipid, and protein metabolism via insulin and AMP-activated protein kinase signaling pathways. Food Funct. 2020;11(2):1798–809.PubMedCrossRef
86.
Zurück zum Zitat Svensson RU, Parker SJ, Eichner LJ, et al. Inhibition of acetyl-CoA carboxylase suppresses fatty acid synthesis and tumor growth of non-small-cell lung cancer in preclinical models. Nat Med. 2016;22(10):1108–19.PubMedPubMedCentralCrossRef Svensson RU, Parker SJ, Eichner LJ, et al. Inhibition of acetyl-CoA carboxylase suppresses fatty acid synthesis and tumor growth of non-small-cell lung cancer in preclinical models. Nat Med. 2016;22(10):1108–19.PubMedPubMedCentralCrossRef
87.
Zurück zum Zitat Siqingaowa SS, Gopalakrishnan V, Taghibiglou C. Sterol regulatory element-binding protein 1 inhibitors decrease pancreatic cancer cell viability and proliferation. Biochem Biophys Res Commun. 2017;488(1):136–40.PubMedCrossRef Siqingaowa SS, Gopalakrishnan V, Taghibiglou C. Sterol regulatory element-binding protein 1 inhibitors decrease pancreatic cancer cell viability and proliferation. Biochem Biophys Res Commun. 2017;488(1):136–40.PubMedCrossRef
88.
Zurück zum Zitat Liu G, Kuang S, Cao R, Wang J, Peng Q, Sun C. Sorafenib kills liver cancer cells by disrupting SCD1-mediated synthesis of monounsaturated fatty acids via the ATP-AMPK-mTOR-SREBP1 signaling pathway. FASEB J. 2019;33(9):10089–103.PubMedCrossRef Liu G, Kuang S, Cao R, Wang J, Peng Q, Sun C. Sorafenib kills liver cancer cells by disrupting SCD1-mediated synthesis of monounsaturated fatty acids via the ATP-AMPK-mTOR-SREBP1 signaling pathway. FASEB J. 2019;33(9):10089–103.PubMedCrossRef
89.
Zurück zum Zitat Gugiatti E, Tenca C, Ravera S, et al. A reversible carnitine palmitoyltransferase (CPT1) inhibitor offsets the proliferation of chronic lymphocytic leukemia cells. Haematologica. 2018;103(11):e531–6.PubMedPubMedCentralCrossRef Gugiatti E, Tenca C, Ravera S, et al. A reversible carnitine palmitoyltransferase (CPT1) inhibitor offsets the proliferation of chronic lymphocytic leukemia cells. Haematologica. 2018;103(11):e531–6.PubMedPubMedCentralCrossRef
90.
Zurück zum Zitat Luo J, Hong Y, Lu Y, et al. Acetyl-CoA carboxylase rewires cancer metabolism to allow cancer cells to survive inhibition of the warburg effect by cetuximab. Cancer Lett. 2017;384:39–49.PubMedCrossRef Luo J, Hong Y, Lu Y, et al. Acetyl-CoA carboxylase rewires cancer metabolism to allow cancer cells to survive inhibition of the warburg effect by cetuximab. Cancer Lett. 2017;384:39–49.PubMedCrossRef
91.
Zurück zum Zitat Talebi A, Dehairs J, Rambow F, et al. Sustained SREBP-1-dependent lipogenesis as a key mediator of resistance to BRAF-targeted therapy. Nat Commun. 2018;9(1):2500.PubMedPubMedCentralCrossRef Talebi A, Dehairs J, Rambow F, et al. Sustained SREBP-1-dependent lipogenesis as a key mediator of resistance to BRAF-targeted therapy. Nat Commun. 2018;9(1):2500.PubMedPubMedCentralCrossRef
Metadaten
Titel
The role of liver kinase B1 in tumor progression through regulation of lipid metabolism
verfasst von
Jialu Geng
Yanghe Zhang
Qingfei Meng
Hang Yan
Yishu Wang
Publikationsdatum
27.07.2022
Verlag
Springer International Publishing
Erschienen in
Clinical and Translational Oncology / Ausgabe 11/2022
Print ISSN: 1699-048X
Elektronische ISSN: 1699-3055
DOI
https://doi.org/10.1007/s12094-022-02863-2

Weitere Artikel der Ausgabe 11/2022

Clinical and Translational Oncology 11/2022 Zur Ausgabe

Alphablocker schützt vor Miktionsproblemen nach der Biopsie

16.05.2024 alpha-1-Rezeptorantagonisten Nachrichten

Nach einer Prostatabiopsie treten häufig Probleme beim Wasserlassen auf. Ob sich das durch den periinterventionellen Einsatz von Alphablockern verhindern lässt, haben australische Mediziner im Zuge einer Metaanalyse untersucht.

Antikörper-Wirkstoff-Konjugat hält solide Tumoren in Schach

16.05.2024 Zielgerichtete Therapie Nachrichten

Trastuzumab deruxtecan scheint auch jenseits von Lungenkrebs gut gegen solide Tumoren mit HER2-Mutationen zu wirken. Dafür sprechen die Daten einer offenen Pan-Tumor-Studie.

Mammakarzinom: Senken Statine das krebsbedingte Sterberisiko?

15.05.2024 Mammakarzinom Nachrichten

Frauen mit lokalem oder metastasiertem Brustkrebs, die Statine einnehmen, haben eine niedrigere krebsspezifische Mortalität als Patientinnen, die dies nicht tun, legen neue Daten aus den USA nahe.

Labor, CT-Anthropometrie zeigen Risiko für Pankreaskrebs

13.05.2024 Pankreaskarzinom Nachrichten

Gerade bei aggressiven Malignomen wie dem duktalen Adenokarzinom des Pankreas könnte Früherkennung die Therapiechancen verbessern. Noch jedoch klafft hier eine Lücke. Ein Studienteam hat einen Weg gesucht, sie zu schließen.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.