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 -- 2674 2023-01-16 20:19:13 |
2 format correction -28 word(s) 2646 2023-01-17 04:42:38 |

Video Upload Options

We provide professional Video Production Services to translate complex research into visually appealing presentations. Would you like to try it?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Jiménez-Cortegana, C.;  Hontecillas-Prieto, L.;  García-Domínguez, D.J.;  Zapata, F.;  Palazón-Carrión, N.;  Sánchez-León, M.L.;  Tami, M.;  Pérez-Pérez, A.;  Sánchez-Jiménez, F.;  Vilariño-García, T.; et al. Leptin and Lymphoma. Encyclopedia. Available online: https://encyclopedia.pub/entry/40250 (accessed on 20 November 2024).
Jiménez-Cortegana C,  Hontecillas-Prieto L,  García-Domínguez DJ,  Zapata F,  Palazón-Carrión N,  Sánchez-León ML, et al. Leptin and Lymphoma. Encyclopedia. Available at: https://encyclopedia.pub/entry/40250. Accessed November 20, 2024.
Jiménez-Cortegana, Carlos, Lourdes Hontecillas-Prieto, Daniel J. García-Domínguez, Fernando Zapata, Natalia Palazón-Carrión, María L. Sánchez-León, Malika Tami, Antonio Pérez-Pérez, Flora Sánchez-Jiménez, Teresa Vilariño-García, et al. "Leptin and Lymphoma" Encyclopedia, https://encyclopedia.pub/entry/40250 (accessed November 20, 2024).
Jiménez-Cortegana, C.,  Hontecillas-Prieto, L.,  García-Domínguez, D.J.,  Zapata, F.,  Palazón-Carrión, N.,  Sánchez-León, M.L.,  Tami, M.,  Pérez-Pérez, A.,  Sánchez-Jiménez, F.,  Vilariño-García, T.,  Cruz-Merino, L.D.L., & Sánchez-Margalet, V. (2023, January 16). Leptin and Lymphoma. In Encyclopedia. https://encyclopedia.pub/entry/40250
Jiménez-Cortegana, Carlos, et al. "Leptin and Lymphoma." Encyclopedia. Web. 16 January, 2023.
Leptin and Lymphoma
Edit

Obesity is a risk factor for many different types of cancer including lymphoma. Since hyperleptinemia is one of the mechanisms of pathophysiology of obesity such as inflammation.

lymphoma obesity leptin

1. Leptin Signaling in Lymphoma

The metabolic abnormalities associated with an excess of adipose tissue include biochemical alterations such as high levels of plasma triglycerides [1] or peripheral insulin resistance, which lead to increased levels of insulin and glucose [2]. Importantly, other factors closely involved in obesity have been described as promoters of many diseases in the last decades, such as the adipokines leptin or adiponectin. Leptin is known to activate and promote the proliferation of monocytes and lymphocytes by activating JAK-STAT, PI3K, and MAPK [3][4]. Leptin signaling also drives the activation of many oncogenic pathways leading to the increased proliferation, epithelial-mesenchymal transition, migration, and invasion of tumor cells [5]. Specifically, leptin signaling pathways can promote lymphomas (Figure 1). Leptin binds its receptor LEPR-b to transduce activation signals into cells via JAK2, which is phosphorylated together with Tyr985, Tyr1077, and Tyr1138. STAT3 proteins bind phospho-Tyr1138 and are phosphorylated and translocated into the nucleus of dimeric units, activating the transcription of their targeting genes and leading to a huge variety of lymphomas, including DLBCL, unclassifiable diseases with features between DLBCL and Burkitt lymphoma, mantle cell (MCL), NK/T-cell (NKTCL), peripheral T-cell (PTCL), anaplastic large cell (ALCL) or intestinal T-cell lymphomas, as well as HL [6][7]. One of its targeting genes, the suppressor of cytokine signaling (SOCS)-3, has been found to be highly expressed in FL and ALCL [8][9]. Similarly, STAT5 binds phospho-Tyr1077 and is translocated into the nucleus after its phosphorylation, thus promoting not only DLBCL, PTCL, MCL, or HL (as STAT3 signaling does) but also γδ-T-cell and lymphoblastic lymphomas [10][11][12][13][14][15].
Figure 1. Leptin signaling pathways that could promote different types of lymphoma. ALCL—anaplastic large cell lymphoma; DLBCL—diffuse large B-cell lymphoma; FL—follicular lymphoma; HL—Hodgkin lymphoma; MCL—mantle cell lymphoma; NKTCL—natural killer/T-cell lymphoma; PTCL—peripheral T cell lymphoma; TCL—T-cell lymphoma.
Moreover, SHP2 binds to phospho-Tyr985 and promotes the activation of the MAPK pathway, although leptin can also activate MAPK signaling independent of SHP2. The protein SHP2 has been associated with ALCL [16][17], whereas MAPK activity impairs outcomes in DLBCL, pediatric-type nodal FL, and plasmablastic lymphoma [18][19][20]. The phosphorylation of JAK2 also promotes the PI3K/AKT/mTOR signaling pathway via IRS activation. The IRS proteins are a family of cytoplasmic adaptor proteins with important roles in cancer [21]. Regarding lymphomas, IRS-1 has been demonstrated to activate anaplastic lymphoma kinase (ALK), which is involved in ALCL [22], and IRS-4 could mediate the mitogenic signaling of LB cells: a murine pre-T-cell lymphoma [23]. The activation of the PI3K/protein kinase B (AKT)/mammalian target of the rapamycin (mTOR) pathway also plays a key role in lymphoma, and many signaling pathway inhibitors have been developed to treat FL, DLBCL, MCL, small lymphocytic, and T-cell NHL [24][25][26][27][28].
At the cellular level, leptin signaling favors Th1 responses by enhancing IL-2, interferon (IFN)-γ synthesis, and inhibiting IL-4 production, which suggests that this adipokine may alter T-cell responses toward a proinflammatory phenotype [29][30]. The recruitment of proinflammatory cytokines by leptin could regulate the production of adhesion molecules, such as the vascular cell adhesion molecule (VCAM)-1 and intercellular cell adhesion molecule (ICAM)-1 [31][32], that have been found to be highly increased in newly diagnosed lymphoma patients and correlate with tumor dissemination, the aggressiveness of the disease, and worse response to treatments [33][34][35][36].
Leptin also induces TNF-α in many settings [37][38][39]. Although its role in cancer remains controversial (as previously explained), TNF-α has been shown to play a key role in the pathogenesis of NHL [40] and may increase the risk of disease together with leptin, especially in FL [41] and DLBCL, through polymorphisms in the TNF rs1800629G>A gene [42]. Additionally, TNF-α levels were higher in lymphoma from children compared with their solid-tumor counterparts [43], which suggests the relevant role of this protein in lymphomas. IL-10 and IFN-γ released by leptin may be implicated in lymphomagenesis since their circulating levels were increased in patients with BMI ≥ 25 kg/m2 compared to individuals with a lower BMI [44]. Although IL-10 may be associated with a higher risk of NHL, especially FL, IFN-γ was not associated with that risk [41]. The risk of lymphoma in patients with a higher BMI could be also increased by the release of IL-6 via leptin signaling [44], but it still needs to be completely elucidated since other studies did not find this association [41]. Of note, blood glucose was suggested as a prognostic biomarker for TCL [45], and the human oocyte testis gene 1, an antigen whose disruption promotes aberrant glucose homeostasis and defective hormone secretion, has been shown to decrease levels of insulin and leptin in TCL-bearing mice [46].

2. Leptin and LEPR Genes in Lymphoma

Leptin has been suggested to promote immune dysfunctions regarding body weight regulation and NHL: mainly DLBCL and FL. Regarding gene expression, lymphomas are mainly characterized by mutations that involve genes, such as B-cell lymphoma (bcl)-2 [47], bcl-6 [48], p15 and p16 [49], p53 [50], or myc [51], which have been widely considered as biomarkers of poor prognosis in those diseases [52][53][54][55][56][57][58]. Specifically, BCL-2 is an antiapoptotic protein that belongs to the BCL-2 family together with other proteins, including (but not limited to) CL-XL and BCL-W, with antiapoptotic properties, as well as the proapoptotic BAX, BAK, or BID proteins [59]. Leptin signaling has been demonstrated to play a key role in B-cell homeostasis through the induction of Bcl-2 [60], which could increase the risk of different pathological conditions. Leptin has demonstrated the ability to inhibit apoptosis and induce cell cycle by elevating Bcl-2 and cyclin D1 in leptin-receptor-deficient (db/db) mice [60]. Similarly, the Bcl-2 protein expression was elevated in db/db mice with diabetes [61][62][63], Which may be predisposed to develop lymphoma [64]. This adipokine also decreased the apoptosis of myocardial cells in rats via bcl-2 [65] and reduced the apoptosis of beta cells at physiological concentrations in vitro by maintaining or up-regulating bcl-2 expression, which could promote non-insulin-dependent diabetes mellitus [66][67]. Additionally, mild maternal protein deprivation during lactation in rat pups could affect thymic homeostasis by increasing the activity of leptin, which improves the levels of BCL-2 and inhibits the apoptosis of thymocytes [68]. In human trophoblasts, leptin also prevents apoptosis when elicited with high temperatures by increasing the BCL-2/BAX ratio [69]. In cancer, the silencing of leptin in HeLa cells, a cervical cancer cell line, has reduced the expression of bcl-2 and, consequently, promotes apoptosis and inhibits cell proliferation, thus suggesting the probable role of leptin in the progression of cervical cancer [70]. Those notions are especially significant since NF-kB, STAT3, PI3K, and AKT pathways are activated in lymphoma cells via leptin/LEPR signaling [71][72][73] and improving bcl-2 expression.
Several studies have analyzed the role of leptin genes in lymphomas (Table 1). Single nucleotide polymorphisms (SNPs) in leptin genes LEP 2548GA and LEP 2548AA have been shown to increase the risk of FL compared with LEP 2548GG [74]. Specifically, genetic polymorphisms in LEP 2548GA have been significantly associated with NHL under the homozygous co-dominant model and additive genetic model in the Caucasian population rather than among Asians after analyzing almost 7000 cases and 8000 controls [75]. The positive associations between LEP 2548GA and the susceptibility of NHL were also found in another study, but without statistically significant differences [76]. Moreover, SNPs in LEP 2548GA have not been suggested to increase the risk of cutaneous T cell lymphomas (TCL) but may be involved in the pharmacogenetic of different treatments for this disease since patients with AG or GG genotypes (with lower plasma leptin levels) could better respond to topical steroids (male patients) and phototherapy (female counterparts) compared with AA patients [77].
Table 1. Leptin/LEPR gene polymorphisms analyzed in lymphomas. NHL—non-Hodgkin lymphoma; DLBCL—diffuse large B-cell lymphoma; FL—follicular lymphoma.
Reference Type of Study Leptin/LEPR Genes Conclusions
[78] Case-control LEP 19AG, LEP 2548GA, LEP 2548AA, and LEPR Q223R Polymorphism in LEP 19AG increased the risk of DLBCL and FL. Genetic interactions in LEPR 223RR, LEP 2548GA, or LEP 2548AA genes also increased the risk of NHL.
[74]. Case-control LEP 2548GA, LEP 2548AA, LEP 19AA, and LEPR 223Q>R Obesity was associated with risk of NHL, especially DLBCL.
The risk of NHL was increased by LEP 2548GA and LEP 2548AA genes and decreased by LEP 19AA, particularly in men younger than in 45 years olds with FL. Conversely, no associations were found between lymphoma risk and LEPR 223Q>R.
[77] Case-control LEP 2548GA Cutaneous T-cell lymphoma patients with leptin genes involving AG or GG genotypes may respond better to topical steroids and phototherapy.
[75] Meta-analysis LEP 2548GA Gene polymorphism may increase the risk of NHL, particularly in the homozygote co-dominant model and the additive genetic model of Caucasian populations.
[79] Meta-analysis LEP 19AG Gene polymorphism was associated with lower NHL risk under the homozygous codominant model, recessive genetic model (especially among the Latin American population), and additive genetic model.
[80] Meta-analysis LEPR Q223R Gene polymorphism did not affect the risk of NHL, although it may be significantly increased in Asian and African individuals.
[76] Meta-analysis LEP 2548GA, and LEP 19AG LEP 2548GA polymorphism increases NHL susceptibility and LEP 19AG is associated with a decreased risk of NHL, especially FL.
[81] Meta-analysis LEP 19AG LEP 19AG may decrease the risk of NHL, especially in Asians, Caucasians, and mixed populations.
[42] Case-control LEPR rs1327118G>C, and LEP rs2167270G>A (LEP 19AG) LEP rs2167270 G>A polymorphism was associated with the decreased risk of DLBCL in the recessive mode models among the Jordanian Arab population.
However, results regarding SNPs in other leptin genes, such as LEP A19G (also known as rs2167270) and its receptor LEPR Q223R, remain inconsistent. Polymorphisms in the LEP A19G gene have been correlated with BMI and an increased risk of DLCL and FL [78], but accumulating evidence from recent years has revealed that SNPs in LEP A19G are associated with a decreased risk of DLBCL [42] and FL [76]. In line with this, meta-analyses have reported that genetic polymorphisms in the LEP A19G gene were associated with a lower risk (or even decrease in the risk) of NHL among Latin American individuals [79] and Asians, Caucasians and mixed populations [81]. Additionally, polymorphisms in the LEP A19G receptor, LEPR Q223R, could not increase the susceptibility of NHL [80]. Other leptin genes, such as LEP 19AA, could decrease that risk [74], whereas the leptin receptor gene rs1327118 G>C has not been associated with susceptibility to the disease [42].
By contrast, the ghrelin GHRL SNP allele for GHRL 4427G>A has been inversely correlated with the risk of NHL, especially DLCL [82]. GHLR and leptin are hormones that play antagonistic roles in controlling energy balance [83] by increasing and decreasing the levels of neuropeptide Y (NPY), respectively [84][85]. NPY is a powerful appetite stimulator that serves as an immune mediator by releasing and inhibiting proinflammatory cytokines [82]. The role of NPY in disease risk and progression remains unclear since it has been found that NPY genes may affect the risk of NHL, especially FL [82], but no significant changes in NPY levels after treatment have been revealed in patients with different types of cancer, including NHL [40].

3. Serum Leptin and LEPR Expression in Lymphoma

Similar to leptin genes, the relationship between the concentration of circulating leptin or LEPR expression and lymphomas has also been studied (Table 2). It has been demonstrated that leptin levels increase the risk of NHL in individuals with BMI ≥ 25 kg/m2 [44]. In addition, the phosphorylation of STAT3 and AKT via JAK2/STAT and PI3K/AKT signaling pathways has led immunohistochemical studies to reveal high expressions of LEPR, p-STAT3, and p-AKT in many DLBCL cases [73]. In line with this notion, leptin has been demonstrated to stimulate the proliferation of DLBCL cells and inhibit apoptosis via the PI3K/AKT signaling pathway in vitro, whereas the pretreatment of DLBCL cells with LEPR-specific siRNA or the inactivation of PI3K/AKT activity depleted these responses [72]. Likewise, leptin has increased the cell viability of CTL MOLT-3 cells by promoting the recruitment and expression of Glut1, and LEPR-siRNA, which inhibited those responses [86].
Table 2. Leptin/LEPR levels or expression analyzed in lymphomas.
Reference Type of Study Leptin/LEPR Levels or Expression Conclusions
[87] Case study All patients: 23 (0–310) pg/mL.
CR patients: 25 (0–310) pg/mL.
PD patients: 21.5 (0–140) pg/mL.
Leptin levels were similar regardless of the response to treatment.
[40] Case-control Article not available * After treatment, BMI, body weight and body fat mass decreased significantly. Also, low leptin levels were found before and after treatment compared with controls.
[88] Case-control Lymphoma patients: 16.4 ± 10.4 ng/mL.
Controls: 10.3 ± 7.6 ng/mL.
Leptin levels were positively correlated with BMI but were not high in lymphoma patients at diagnosis.
[89] Case-control Patients: 6.0 ± 6.31 ng/mL.
Controls: 5.9 ± 7.3 ng/mL.
There was no association between leptin levels and NHL in children.
[90] Case-control Patients: 8.2 ± 7.26 ng/mL.
Controls: 7.5 ± 8.3 ng/mL.
There was no association between leptin levels and HL in children.
[72] Case study High LEPR expression in 39.8% of DLBCL patients LEPR overexpression could be associated with DLBCL carcinogenesis via PI3K/AKT pathway. Also, leptin/LEPR signaling promoted the proliferation of DLBCL cells in vitro.
[91] Case-control Pre-treatment:
5.3 ± 1.56 ng/mL.
Post-treatment: 9.8 ± 2.7 ng/mL.
Controls: 6.7 ± 1.2 ng/mL.
Leptin levels were significantly lower in patients than in controls and increased in patients who achieved remission.
[41] Case-control Patients: 8.5 (3.8–17.1) ng/mL.
Controls: 10.6 (5.2–21.8) ng/mL.
Serum leptin levels were significantly associated with NHL risk at diagnosis, but predicted a lower risk of FL.
[73] Case-control High LEPR expression in 45% of DLBCL patients LEPR may promote JAK/STAT and PI3K/AKT signaling pathways and induce the phosphorylation of STAT3 and AKT, which may be involved in the prognosis of DLBCL.
[86] Cases Higher LEPR expression in tissues of T-cell lymphoma patients (58.3%) and in all cell lines, especially in MOLT-3 and Jurkat cell lines LEPR overexpression was positively correlated with Glut1 expression.
TCL MOLT-3 cell line demonstrated that leptin stimulated cell glucose uptake via promoting the recruitment and expression of Glut1.
[44] Case-control Patients: 4182.30 ± 246.95 pg/mL.
Controls: 4782.00  ±  193.65 pg/mL
Leptin levels were significantly higher in women than in men and in obese patients compared with their non-obese counterparts, which increased the risk of NHL.
NHL—non-Hodgkin lymphoma; DLBCL—diffuse large B-cell lymphoma; FL—follicular lymphoma. * Article not available to check circulating leptin levels. The rest of the information was taken from the abstract.
By contrast, it has also been shown that leptin levels not only undergo slight changes after treatment in NHL adult patients [40] but also could be negatively correlated with the international prognostic score in HL and with the international prognostic index in NHL [88], suggesting a paradoxical role of leptin that has been previously explained not only in cancer [92] but also in other settings [93][94][95]. Also, Bertolini et al. (1999) studied patients with NHL (mainly DLBCL and FL but also other types of lymphomas such as MCL, PTCL, ALCL, large granular NK-cell lymphoma, and extranodal marginal zone lymphoma of MALT), whose leptin levels were not only similar regardless of the outcome but were also not correlated with age, gender, or even-free survival [87].

4. Linking Leptin, Lymphoma, and Obesity

Most of the studies have analyzed the relationship between leptin and lymphoma, obesity and lymphoma, or obesity and leptin. Therefore, only a few studies have analyzed the possible associations among leptin, lymphoma, and obesity. Recently, leptin has been positively associated with BMI and NHL risk [44]. Also, patients who survived the Burkitt type, non-Burkitt, and lymphoblastic lymphomas not only had low leptin levels but also a normal/low BMI (19.5 ± 3.4 kg/m2) [96]. On the other hand, leptin levels have been positively correlated with BMI but not associated with lymphoma risk [88]. Similarly, relationships between leptin or BMI with HL or NHL were not found in pediatric patients [90]. A BMI ranging from underweight to healthy values in children newly diagnosed with HL or NHL has been positively correlated with leptin. Thus, leptin levels were low in those patients at diagnosis [89][91] but may significantly increase after remission and predict the response to treatment or progressive disease [91]. Regarding SNP, it has been found that leptin gene polymorphisms were independent of BMI and did not alter the risk of NHL [74].

References

  1. van de Woestijne, A.P.; Monajemi, H.; Kalkhoven, E.; Visseren, F.L. Adipose tissue dysfunction and hypertriglyceridemia: Mechanisms and management. Obes. Rev. 2011, 12, 829–840.
  2. Gastaldelli, A.; Gaggini, M.; DeFronzo, R.A. Role of Adipose Tissue Insulin Resistance in the Natural History of Type 2 Diabetes: Results From the San Antonio Metabolism Study. Diabetes 2017, 66, 815–822.
  3. Sanchez-Margalet, V.; Martin-Romero, C. Human leptin signaling in human peripheral blood mononuclear cells: Activation of the JAK-STAT pathway. Cell Immunol. 2001, 211, 30–36.
  4. Martin-Romero, C.; Sanchez-Margalet, V. Human leptin activates PI3K and MAPK pathways in human peripheral blood mononuclear cells: Possible role of Sam68. Cell Immunol. 2001, 212, 83–91.
  5. Ghasemi, A.; Saeidi, J.; Azimi-Nejad, M.; Hashemy, S.I. Leptin-induced signaling pathways in cancer cell migration and invasion. Cell Oncol. 2019, 42, 243–260.
  6. Ding, B.B.; Yu, J.J.; Yu, R.Y.; Mendez, L.M.; Shaknovich, R.; Zhang, Y.; Cattoretti, G.; Ye, B.H. Constitutively activated STAT3 promotes cell proliferation and survival in the activated B-cell subtype of diffuse large B-cell lymphomas. Blood 2008, 111, 1515–1523.
  7. Zhu, F.; Wang, K.B.; Rui, L. STAT3 Activation and Oncogenesis in Lymphoma. Cancers 2019, 12, 19.
  8. Krishnadasan, R.; Bifulco, C.; Kim, J.; Rodov, S.; Zieske, A.W.; Vanasse, G.J. Overexpression of SOCS3 is associated with decreased survival in a cohort of patients with de novo follicular lymphoma. Br. J. Haematol. 2006, 135, 72–75.
  9. Cho-Vega, J.H.; Rassidakis, G.Z.; Amin, H.M.; Tsioli, P.; Spurgers, K.; Remache, Y.K.; Vega, F.; Goy, A.H.; Gilles, F.; Medeiros, L.J. Suppressor of cytokine signaling 3 expression in anaplastic large cell lymphoma. Leukemia 2004, 18, 1872–1878.
  10. Maurer, B.; Nivarthi, H.; Wingelhofer, B.; Pham, H.T.T.; Schlederer, M.; Suske, T.; Grausenburger, R.; Schiefer, A.I.; Prchal-Murphy, M.; Chen, D.; et al. High activation of STAT5A drives peripheral T-cell lymphoma and leukemia. Haematologica 2020, 105, 435–447.
  11. Kucuk, C.; Jiang, B.; Hu, X.; Zhang, W.; Chan, J.K.; Xiao, W.; Lack, N.; Alkan, C.; Williams, J.C.; Avery, K.N.; et al. Activating mutations of STAT5B and STAT3 in lymphomas derived from gammadelta-T or NK cells. Nat. Commun. 2015, 6, 1–12.
  12. Martini, M.; Hohaus, S.; Petrucci, G.; Cenci, T.; Pierconti, F.; Massini, G.; Teofili, L.; Leone, G.; Larocca, L.M. Phosphorylated STAT5 represents a new possible prognostic marker in Hodgkin lymphoma. Am. J. Clin. Pathol. 2008, 129, 472–477.
  13. Scheeren, F.A.; Diehl, S.A.; Smit, L.A.; Beaumont, T.; Naspetti, M.; Bende, R.J.; Blom, B.; Karube, K.; Ohshima, K.; van Noesel, C.J.; et al. IL-21 is expressed in Hodgkin lymphoma and activates STAT5: Evidence that activated STAT5 is required for Hodgkin lymphomagenesis. Blood 2008, 111, 4706–4715.
  14. Kelly, J.A.; Spolski, R.; Kovanen, P.E.; Suzuki, T.; Bollenbacher, J.; Pise-Masison, C.A.; Radonovich, M.F.; Lee, S.; Jenkins, N.A.; Copeland, N.G.; et al. Stat5 synergizes with T cell receptor/antigen stimulation in the development of lymphoblastic lymphoma. J. Exp. Med. 2003, 198, 79–89.
  15. Shipp, M.A.; Ross, K.N.; Tamayo, P.; Weng, A.P.; Kutok, J.L.; Aguiar, R.C.; Gaasenbeek, M.; Angelo, M.; Reich, M.; Pinkus, G.S.; et al. Diffuse large B-cell lymphoma outcome prediction by gene-expression profiling and supervised machine learning. Nat. Med. 2002, 8, 68–74.
  16. Voena, C.; Conte, C.; Ambrogio, C.; Boeri Erba, E.; Boccalatte, F.; Mohammed, S.; Jensen, O.N.; Palestro, G.; Inghirami, G.; Chiarle, R. The tyrosine phosphatase Shp2 interacts with NPM-ALK and regulates anaplastic lymphoma cell growth and migration. Cancer Res. 2007, 67, 4278–4286.
  17. Karaca Atabay, E.; Mecca, C.; Wang, Q.; Ambrogio, C.; Mota, I.; Prokoph, N.; Mura, G.; Martinengo, C.; Patrucco, E.; Leonardi, G.; et al. Tyrosine phosphatases regulate resistance to ALK inhibitors in ALK+ anaplastic large cell lymphoma. Blood 2022, 139, 717–731.
  18. Vega, G.G.; Aviles-Salas, A.; Chalapud, J.R.; Martinez-Paniagua, M.; Pelayo, R.; Mayani, H.; Hernandez-Pando, R.; Martinez-Maza, O.; Huerta-Yepez, S.; Bonavida, B.; et al. P38 MAPK expression and activation predicts failure of response to CHOP in patients with Diffuse Large B-Cell Lymphoma. BMC Cancer 2015, 15, 722.
  19. Louissaint, A., Jr.; Schafernak, K.T.; Geyer, J.T.; Kovach, A.E.; Ghandi, M.; Gratzinger, D.; Roth, C.G.; Paxton, C.N.; Kim, S.; Namgyal, C.; et al. Pediatric-type nodal follicular lymphoma: A biologically distinct lymphoma with frequent MAPK pathway mutations. Blood 2016, 128, 1093–1100.
  20. Ramis-Zaldivar, J.E.; Gonzalez-Farre, B.; Nicolae, A.; Pack, S.; Clot, G.; Nadeu, F.; Mottok, A.; Horn, H.; Song, J.Y.; Fu, K.; et al. MAPK and JAK-STAT pathways dysregulation in plasmablastic lymphoma. Haematologica 2021, 106, 2682–2693.
  21. Shaw, L.M. The insulin receptor substrate (IRS) proteins: At the intersection of metabolism and cancer. Cell Cycle 2011, 10, 1750–1756.
  22. Kuo, A.H.; Stoica, G.E.; Riegel, A.T.; Wellstein, A. Recruitment of insulin receptor substrate-1 and activation of NF-kappaB essential for midkine growth signaling through anaplastic lymphoma kinase. Oncogene 2007, 26, 859–869.
  23. Urso, B.; Ilondo, M.M.; Holst, P.A.; Christoffersen, C.T.; Ouwens, M.; Giorgetti, S.; Van Obberghen, E.; Naor, D.; Tornqvist, H.; De Meyts, P. IRS-4 mediated mitogenic signalling by insulin and growth hormone in LB cells, a murine T-cell lymphoma devoid of IGF-I receptors. Cell. Signal. 2003, 15, 385–394.
  24. Pongas, G.; Cheson, B.D. PI3K signaling pathway in normal B cells and indolent B-cell malignancies. Semin. Oncol. 2016, 43, 647–654.
  25. Cui, W.; Cai, Y.; Wang, W.; Liu, Z.; Wei, P.; Bi, R.; Chen, W.; Sun, M.; Zhou, X. Frequent copy number variations of PI3K/AKT pathway and aberrant protein expressions of PI3K subunits are associated with inferior survival in diffuse large B cell lymphoma. J. Transl. Med. 2014, 12, 10.
  26. Iyengar, S.; Clear, A.; Bodor, C.; Maharaj, L.; Lee, A.; Calaminici, M.; Matthews, J.; Iqbal, S.; Auer, R.; Gribben, J.; et al. P110alpha-mediated constitutive PI3K signaling limits the efficacy of p110delta-selective inhibition in mantle cell lymphoma, particularly with multiple relapse. Blood 2013, 121, 2274–2284.
  27. Shah, A.; Barrientos, J.C. Oral PI3K-delta, gamma Inhibitor for the Management of People with Chronic Lymphocytic Leukemia and Small Lymphocytic Lymphoma: A Narrative Review on Duvelisib. OncoTargets Ther. 2021, 14, 2109–2119.
  28. Huang, D.; Song, T.L.; Nairismagi, M.L.; Laurensia, Y.; Pang, W.L.; Zhe, D.C.M.; Wong, E.K.Y.; Wijaya, G.G.; Tan, J.; Tan, S.H.; et al. Evaluation of the PIK3 pathway in peripheral T-cell lymphoma and NK/T-cell lymphoma. Br. J. Haematol. 2020, 189, 731–744.
  29. Sanchez-Margalet, V.; Martin-Romero, C.; Santos-Alvarez, J.; Goberna, R.; Najib, S.; Gonzalez-Yanes, C. Role of leptin as an immunomodulator of blood mononuclear cells: Mechanisms of action. Clin. Exp. Immunol. 2003, 133, 11–19.
  30. Raso, G.M.; Pacilio, M.; Esposito, E.; Coppola, A.; Di Carlo, R.; Meli, R. Leptin potentiates IFN-gamma-induced expression of nitric oxide synthase and cyclo-oxygenase-2 in murine macrophage J774A.1. Br. J. Pharmacol. 2002, 137, 799–804.
  31. Conde, J.; Scotece, M.; Lopez, V.; Gomez, R.; Lago, F.; Pino, J.; Gomez-Reino, J.J.; Gualillo, O. Adiponectin and leptin induce VCAM-1 expression in human and murine chondrocytes. PLoS ONE 2012, 7, e52533.
  32. Suzukawa, M.; Koketsu, R.; Baba, S.; Igarashi, S.; Nagase, H.; Yamaguchi, M.; Matsutani, N.; Kawamura, M.; Shoji, S.; Hebisawa, A.; et al. Leptin enhances ICAM-1 expression, induces migration and cytokine synthesis, and prolongs survival of human airway epithelial cells. Am. J. Physiol.-Lung Cell. Mol. Physiol. 2015, 309, L801-811.
  33. Terol, M.J.; Lopez-Guillermo, A.; Bosch, F.; Villamor, N.; Cid, M.C.; Rozman, C.; Campo, E.; Montserrat, E. Expression of the adhesion molecule ICAM-1 in non-Hodgkin’s lymphoma: Relationship with tumor dissemination and prognostic importance. J. Clin. Oncol. 1998, 16, 35–40.
  34. Jacob, M.C.; Agrawal, S.; Chaperot, L.; Giroux, C.; Gressin, R.; Le Marc’Hadour, F.; Favre, M.; Sotto, J.J.; Bensa, J.C.; Plumas, J. Quantification of cellular adhesion molecules on malignant B cells from non-Hodgkin’s lymphoma. Leukemia 1999, 13, 1428–1433.
  35. Liu, Y.; Gu, J.J.; Yang, L.; Tsai, P.C.; Guo, Y.; Xue, K.; Xia, Z.; Liu, X.; Lv, F.; Cao, J.; et al. The adhesion molecule ICAM-1 in diffuse large B-cell lymphoma post-rituximab era: Relationship with prognostic importance and rituximab resistance. Aging 2020, 13, 181–193.
  36. Syrigos, K.N.; Salgami, E.; Karayiannakis, A.J.; Katirtzoglou, N.; Sekara, E.; Roussou, P. Prognostic significance of soluble adhesion molecules in Hodgkin’s disease. Anticancer Res. 2004, 24, 1243–1247.
  37. Agrawal, S.; Gollapudi, S.; Su, H.; Gupta, S. Leptin activates human B cells to secrete TNF-alpha, IL-6, and IL-10 via JAK2/STAT3 and p38MAPK/ERK1/2 signaling pathway. J. Clin. Immunol. 2011, 31, 472–478.
  38. Lee, S.M.; Choi, H.J.; Oh, C.H.; Oh, J.W.; Han, J.S. Leptin increases TNF-alpha expression and production through phospholipase D1 in Raw 264.7 cells. PLoS ONE 2014, 9, e102373.
  39. Ziegler, J.F.; Bottcher, C.; Letizia, M.; Yerinde, C.; Wu, H.; Freise, I.; Rodriguez-Sillke, Y.; Stoyanova, A.K.; Kreis, M.E.; Asbach, P.; et al. Leptin induces TNFalpha-dependent inflammation in acquired generalized lipodystrophy and combined Crohn’s disease. Nat. Commun. 2019, 10, 1–11.
  40. Kowalczuk, A.; Wiecek, A.; Franek, E.; Kokot, F. Plasma concentration of leptin, neuropeptide Y and tumor necrosis factor alpha in patients with cancers, before and after radio- and chemotherapy. Pol. Arch. Med. Wewn. 2001, 106, 657–668.
  41. Conroy, S.M.; Maskarinec, G.; Morimoto, Y.; Franke, A.A.; Cooney, R.V.; Wilkens, L.R.; Goodman, M.T.; Hernadez, B.Y.; Le Marchand, L.; Henderson, B.E.; et al. Non-hodgkin lymphoma and circulating markers of inflammation and adiposity in a nested case-control study: The multiethnic cohort. Cancer Epidemiol. Biomark. Prev. 2013, 22, 337–347.
  42. Al-Khatib, S.M.; Abdo, N.; Al-Eitan, L.N.; Al-Mistarehi, A.W.; Zahran, D.J.; Kewan, T.Z. LTA, LEP, and TNF-a Gene Polymorphisms are Associated with Susceptibility and Overall Survival of Diffuse Large B-Cell lymphoma in an Arab Population: A Case-Control Study. Asian Pac. J. Cancer Prev. 2020, 21, 2783–2791.
  43. Barbosa-Cortes, L.; Klunder-Klunder, M.; Lopez-Alarcon, M.; Marquez, H.R.; Lopez-Aguilar, E.; Tapia-Marcial, A. Nutritional status and cytokine concentration during chemotherapy in Mexican children: A longitudinal analysis. Nutrition 2019, 57, 46–51.
  44. El Demerdash, D.M.; Tawfik, N.M.; Elazab, R.; El Sissy, M.H. The Association of Pre-diagnostic Inflammatory Markers and Adipokines and the Risk of Non-Hodgkin Lymphoma Development in Egypt. Indian J. Hematol. Blood Transfus. 2021, 37, 76–81.
  45. Cai, Q.; Luo, X.; Liang, Y.; Rao, H.; Fang, X.; Jiang, W.; Lin, T.; Lin, T.; Huang, H. Fasting blood glucose is a novel prognostic indicator for extranodal natural killer/T-cell lymphoma, nasal type. Br. J. Cancer 2013, 108, 380–386.
  46. Wang, G.; Li, R.; Yang, Y.; Cai, L.; Ding, S.; Xu, T.; Han, M.; Wu, X. Disruption of the Golgi protein Otg1 gene causes defective hormone secretion and aberrant glucose homeostasis in mice. Cell Biosci. 2016, 6, 1–10.
  47. Lai, R.; Arber, D.A.; Chang, K.L.; Wilson, C.S.; Weiss, L.M. Frequency of bcl-2 expression in non-Hodgkin’s lymphoma: A study of 778 cases with comparison of marginal zone lymphoma and monocytoid B-cell hyperplasia. Mod. Pathol. 1998, 11, 864–869.
  48. Wagner, S.D.; Ahearne, M.; Ko Ferrigno, P. The role of BCL6 in lymphomas and routes to therapy. Br. J. Haematol. 2011, 152, 3–12.
  49. Child, F.J.; Scarisbrick, J.J.; Calonje, E.; Orchard, G.; Russell-Jones, R.; Whittaker, S.J. Inactivation of tumor suppressor genes p15(INK4b) and p16(INK4a) in primary cutaneous B cell lymphoma. J. Investig. Dermatol. 2002, 118, 941–948.
  50. Wang, X.J.; Medeiros, L.J.; Bueso-Ramos, C.E.; Tang, G.; Wang, S.; Oki, Y.; Desai, P.; Khoury, J.D.; Miranda, R.N.; Tang, Z.; et al. P53 expression correlates with poorer survival and augments the negative prognostic effect of MYC rearrangement, expression or concurrent MYC/BCL2 expression in diffuse large B-cell lymphoma. Mod. Pathol. 2017, 30, 194–203.
  51. Xia, Y.; Zhang, X. The Spectrum of MYC Alterations in Diffuse Large B-Cell Lymphoma. Acta Haematol. 2020, 143, 520–528.
  52. Sup, S.J.; Alemany, C.A.; Pohlman, B.; Elson, P.; Malhi, S.; Thakkar, S.; Steinle, R.; Hsi, E.D. Expression of bcl-2 in classical Hodgkin’s lymphoma: An independent predictor of poor outcome. J. Clin. Oncol. 2005, 23, 3773–3779.
  53. Mahmoud, H.M.; El-Sakhawy, Y.N. Significance of Bcl-2 and Bcl-6 immunostaining in B-Non Hodgkin’s lymphoma. Hematol. Rep. 2011, 3, e26.
  54. Xia, B.; Zhang, L.; Guo, S.Q.; Li, X.W.; Qu, F.L.; Zhao, H.F.; Zhang, L.Y.; Sun, B.C.; You, J.; Zhang, Y.Z. Coexpression of MYC and BCL-2 predicts prognosis in primary gastrointestinal diffuse large B-cell lymphoma. World J. Gastroenterol. 2015, 21, 2433–2442.
  55. Patrascu, A.M.; Rotaru, I.; Olar, L.; Patrascu, S.; Ghilusi, M.C.; NeamTu, S.D.; Nacea, J.G.; Gluhovschi, A. The prognostic role of Bcl-2, Ki67, c-MYC and p53 in diffuse large B-cell lymphoma. Rom. J. Morphol. Embryol. 2017, 58, 837–843.
  56. Li, L.; Zhang, X.; Zhang, T.; Song, Z.; Hu, G.; Li, W.; Li, L.; Qiu, L.; Qian, Z.; Zhou, S.; et al. Prognostic Significance of BCL-2 and BCL-6 Expression in MYC-positive DLBCL. Clin. Lymphoma Myeloma Leuk. 2018, 18, e381–e389.
  57. Shi, C.L.; Zhang, X.Y.; Li, Y.; Song, L.L.; Wang, L. Correlations of mouse lymphoma xenografts with the expressions of MMP-9 and Bcl-2. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 1176–1183.
  58. Pileri, A.; Agostinelli, C.; Bertuzzi, C.; Grandi, V.; Maio, V.; Lastrucci, I.; Santucci, M.; Pimpinelli, N. Prognostic significance of Bcl-2 expression in primary cutaneous B-cell lymphoma: A reappraisal. Ital. J. Dermatol. Venerol. 2021, 156, 642–649.
  59. Kale, J.; Osterlund, E.J.; Andrews, D.W. BCL-2 family proteins: Changing partners in the dance towards death. Cell Death Differ. 2018, 25, 65–80.
  60. Lam, Q.L.; Wang, S.; Ko, O.K.; Kincade, P.W.; Lu, L. Leptin signaling maintains B-cell homeostasis via induction of Bcl-2 and Cyclin D1. Proc. Natl. Acad. Sci. USA 2010, 107, 13812–13817.
  61. Michurina, S.V.; Ishchenko, I.Y.; Arkhipov, S.A.; Cherepanova, M.A.; Vasendin, D.V.; Zavjalov, E.L. Apoptosis in the liver of male db/db mice during the development of obesity and type 2 diabetes. Vavilov J. Genet. Breed. 2020, 24, 435–440.
  62. Plante, E.; Menaouar, A.; Danalache, B.A.; Yip, D.; Broderick, T.L.; Chiasson, J.L.; Jankowski, M.; Gutkowska, J. Oxytocin treatment prevents the cardiomyopathy observed in obese diabetic male db/db mice. Endocrinology 2015, 156, 1416–1428.
  63. Chen, W.; Sun, Q.; Ju, J.; Chen, W.; Zhao, X.; Zhang, Y.; Yang, Y. Effect of Astragalus Polysaccharides on Cardiac Dysfunction in db/db Mice with Respect to Oxidant Stress. Biomed. Res. Int. 2018, 2018, 8359013.
  64. Sataranatarajan, K.; Ikeno, Y.; Bokov, A.; Feliers, D.; Yalamanchili, H.; Lee, H.J.; Mariappan, M.M.; Tabatabai-Mir, H.; Diaz, V.; Prasad, S.; et al. Rapamycin Increases Mortality in db/db Mice, a Mouse Model of Type 2 Diabetes. J. Gerontol. A Biol. Sci. Med. Sci. 2016, 71, 850–857.
  65. Wu, D.D.; Wu, X.H.; Zhang, L.N. Effect of leptin on expression of calpain-1 and Bcl-2 and apoptosis in myocardial tissue of neonatal rats after asphyxia. Zhongguo Dang dai er ke za zhi 2016, 18, 1044–1049.
  66. Shimabukuro, M.; Wang, M.Y.; Zhou, Y.T.; Newgard, C.B.; Unger, R.H. Protection against lipoapoptosis of beta cells through leptin-dependent maintenance of Bcl-2 expression. Proc. Natl. Acad. Sci. USA 1998, 95, 9558–9561.
  67. Brown, J.E.; Dunmore, S.J. Leptin decreases apoptosis and alters BCL-2: Bax ratio in clonal rodent pancreatic beta-cells. Diabetes/Metab. Res. Rev. 2007, 23, 497–502.
  68. da Silva, S.V.; Salama, C.; Renovato-Martins, M.; Helal-Neto, E.; Citelli, M.; Savino, W.; Barja-Fidalgo, C. Increased leptin response and inhibition of apoptosis in thymocytes of young rats offspring from protein deprived dams during lactation. PLoS ONE 2013, 8, e64220.
  69. Perez-Perez, A.; Toro, A.R.; Vilarino-Garcia, T.; Guadix, P.; Maymo, J.L.; Duenas, J.L.; Varone, C.L.; Sanchez-Margalet, V. Leptin reduces apoptosis triggered by high temperature in human placental villous explants: The role of the p53 pathway. Placenta 2016, 42, 106–113.
  70. Yuan, Y.; Zhang, J.; Cai, L.; Ding, C.; Wang, X.; Chen, H.; Wang, X.; Yan, J.; Lu, J. Leptin induces cell proliferation and reduces cell apoptosis by activating c-myc in cervical cancer. Oncol. Rep. 2013, 29, 2291–2296.
  71. Han, S.S.; Yun, H.; Son, D.J.; Tompkins, V.S.; Peng, L.; Chung, S.T.; Kim, J.S.; Park, E.S.; Janz, S. NF-kappaB/STAT3/PI3K signaling crosstalk in iMyc E mu B lymphoma. Mol. Cancer 2010, 9, 97.
  72. Uddin, S.; Bu, R.; Ahmed, M.; Hussain, A.R.; Ajarim, D.; Al-Dayel, F.; Bavi, P.; Al-kuraya, K.S. Leptin receptor expression and its association with PI3K/AKT signaling pathway in diffuse large B-cell lymphoma. Leuk. Lymphoma 2010, 51, 1305–1314.
  73. Lin, S.; Li, Y.; Xing, X.M.; Ran, W.W. Expression and significance of leptin receptor, p-STAT3 and p-AKT in diffuse large B-cell lymphoma. Acta Histochem. 2014, 116, 126–130.
  74. Willett, E.V.; Skibola, C.F.; Adamson, P.; Skibola, D.R.; Morgan, G.J.; Smith, M.T.; Roman, E. Non-Hodgkin’s lymphoma, obesity and energy homeostasis polymorphisms. Br. J. Cancer 2005, 93, 811–816.
  75. Yang, Y.; Liu, P.; Guo, F.; Liu, R.; Yang, Y.; Huang, C.; Shu, H.; Gong, J.; Cai, M. Genetic G2548A polymorphism of leptin gene and risk of cancer: A meta-analysis of 6860 cases and 7956 controls. J. BUON 2014, 19, 1096–1104.
  76. Lin, H.Y.; Shi, H.; Li, C.Y.; Chen, Q.C.; Huang, T.B.; Liu, P.C.; Lou, L.M. LEP and LEPR polymorphisms in non-Hodgkin lymphoma risk: A systematic review and pooled analysis. J. BUON 2015, 20, 261–268.
  77. Vasku, V.; Vasku, A.; Vasku, J.B. Pharmacogenetic contribution of leptin gene polymorphism in cutaneous T-cell lymphoma. Int. J. Clin. Exp. Pathol. 2009, 2, 163–168.
  78. Skibola, C.F.; Holly, E.A.; Forrest, M.S.; Hubbard, A.; Bracci, P.M.; Skibola, D.R.; Hegedus, C.; Smith, M.T. Body mass index, leptin and leptin receptor polymorphisms, and non-hodgkin lymphoma. Cancer Epidemiol. Biomark. Prev. 2004, 13, 779–786.
  79. Liu, P.; Shi, H.; Huang, C.; Shu, H.; Liu, R.; Yang, Y.; Gong, J.; Yang, Y.; Cai, M. Association of LEP A19G polymorphism with cancer risk: A systematic review and pooled analysis. Tumour Biol. 2014, 35, 8133–8141.
  80. Liu, P.; Shi, H.; Liu, R.; Yang, Y.; Yang, Y.; Huang, C.; Shu, H.; Gong, J.; Cai, M. Lack of association between LEPR Q223R polymorphisms and cancer susceptibility: Evidence from a meta-analysis. J. BUON 2014, 19, 855–862.
  81. Yang, J.; Zhong, Z.; Tang, W.; Chen, J. Leptin rs2167270 G > A (G19A) polymorphism may decrease the risk of cancer: A case-control study and meta-analysis involving 19 989 subjects. J. Cell. Biochem. 2019, 120, 10998–11007.
  82. Skibola, D.R.; Smith, M.T.; Bracci, P.M.; Hubbard, A.E.; Agana, L.; Chi, S.; Holly, E.A. Polymorphisms in ghrelin and neuropeptide Y genes are associated with non-Hodgkin lymphoma. Cancer Epidemiol. Biomark. Prev. 2005, 14, 1251–1256.
  83. Cui, H.; Lopez, M.; Rahmouni, K. The cellular and molecular bases of leptin and ghrelin resistance in obesity. Nat. Rev. Endocrinol. 2017, 13, 338–351.
  84. Wang, Q.; Bing, C.; Al-Barazanji, K.; Mossakowaska, D.E.; Wang, X.M.; McBay, D.L.; Neville, W.A.; Taddayon, M.; Pickavance, L.; Dryden, S.; et al. Interactions between leptin and hypothalamic neuropeptide Y neurons in the control of food intake and energy homeostasis in the rat. Diabetes 1997, 46, 335–341.
  85. Goto, M.; Arima, H.; Watanabe, M.; Hayashi, M.; Banno, R.; Sato, I.; Nagasaki, H.; Oiso, Y. Ghrelin increases neuropeptide Y and agouti-related peptide gene expression in the arcuate nucleus in rat hypothalamic organotypic cultures. Endocrinology 2006, 147, 5102–5109.
  86. Han, T.J.; Xu, H.Z.; Li, J.S.; Geng, L.Y.; Li, X.Y.; Zhou, X.X.; Wang, X. Leptin and its receptor in glucose metabolism of T-cell lymphoma. Oncol. Lett. 2018, 16, 5838–5846.
  87. Bertolini, F.; Paolucci, M.; Peccatori, F.; Cinieri, S.; Agazzi, A.; Ferrucci, P.F.; Cocorocchio, E.; Goldhirsch, A.; Martinelli, G. Angiogenic growth factors and endostatin in non-Hodgkin’s lymphoma. Br. J. Haematol. 1999, 106, 504–509.
  88. Pamuk, G.E.; Demir, M.; Harmandar, F.; Yesil, Y.; Turgut, B.; Vural, O. Leptin and resistin levels in serum of patients with hematologic malignancies: Correlation with clinical characteristics. Exp. Oncol. 2006, 28, 241–244.
  89. Petridou, E.T.; Sergentanis, T.N.; Dessypris, N.; Vlachantoni, I.T.; Tseleni-Balafouta, S.; Pourtsidis, A.; Moschovi, M.; Polychronopoulou, S.; Athanasiadou-Piperopoulou, F.; Kalmanti, M.; et al. Serum adiponectin as a predictor of childhood non-Hodgkin’s lymphoma: A nationwide case-control study. J. Clin. Oncol. 2009, 27, 5049–5055.
  90. Petridou, E.T.; Dessypris, N.; Panagopoulou, P.; Sergentanis, T.N.; Mentis, A.F.; Pourtsidis, A.; Polychronopoulou, S.; Kalmanti, M.; Athanasiadou-Piperopoulou, F.; Moschovi, M. Adipocytokines in relation to Hodgkin lymphoma in children. Pediatr. Blood Cancer 2010, 54, 311–315.
  91. Okur, F.V.; Karadeniz, C.; Buyukpamukcu, M.; Oguz, A.; Yucel, A.; Cinaz, P.; Emir, S.; Varan, A. Clinical significance of serum vascular endothelial growth factor, endostatin, and leptin levels in children with lymphoma. Pediatr. Blood Cancer 2010, 55, 1272–1277.
  92. Jimenez-Cortegana, C.; Lopez-Saavedra, A.; Sanchez-Jimenez, F.; Perez-Perez, A.; Castineiras, J.; Virizuela-Echaburu, J.A.; de la Cruz-Merino, L.; Sanchez-Margalet, V. Leptin, Both Bad and Good Actor in Cancer. Biomolecules 2021, 11, 913.
  93. Head, G.A. The oestrogen-leptin paradox. J. Physiol. 2015, 593, 1523.
  94. Ren, J. Lessons from the leptin paradox in cardiac regulation--too much versus too little. J. Physiol. 2005, 565, 347.
  95. Zhao, S.; Kusminski, C.M.; Scherer, P.E. Adiponectin, Leptin and Cardiovascular Disorders. Circ. Res. 2021, 128, 136–149.
  96. Yaris, N.; Sozen, E.; Erduran, E.; Okten, A.; Orem, A.; Cakirbay, H. Bone mineral metabolism and its relationship to leptin levels in survivors of childhood leukemia and lymphoma. Pediatr. Hematol. Oncol. 2005, 22, 489–498.
More
Information
Subjects: Oncology
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: 548
Revisions: 2 times (View History)
Update Date: 17 Jan 2023
1000/1000
ScholarVision Creations