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 -- 2944 2023-04-20 21:04:09 |
2 Reference format revised. Meta information modification 2944 2023-04-23 09:40:24 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Villegas-Vázquez, E.Y.; Quintas-Granados, L.I.; Cortés, H.; González-Del Carmen, M.; Leyva-Gómez, G.; Rodríguez-Morales, M.; Bustamante-Montes, L.P.; Silva-Adaya, D.; Pérez-Plasencia, C.; Jacobo-Herrera, N.; et al. Lithium as a Promising Anticancer Agent. Encyclopedia. Available online: https://encyclopedia.pub/entry/43305 (accessed on 19 August 2024).
Villegas-Vázquez EY, Quintas-Granados LI, Cortés H, González-Del Carmen M, Leyva-Gómez G, Rodríguez-Morales M, et al. Lithium as a Promising Anticancer Agent. Encyclopedia. Available at: https://encyclopedia.pub/entry/43305. Accessed August 19, 2024.
Villegas-Vázquez, Edgar Yebrán, Laura Itzel Quintas-Granados, Hernán Cortés, Manuel González-Del Carmen, Gerardo Leyva-Gómez, Miguel Rodríguez-Morales, Lilia Patricia Bustamante-Montes, Daniela Silva-Adaya, Carlos Pérez-Plasencia, Nadia Jacobo-Herrera, et al. "Lithium as a Promising Anticancer Agent" Encyclopedia, https://encyclopedia.pub/entry/43305 (accessed August 19, 2024).
Villegas-Vázquez, E.Y., Quintas-Granados, L.I., Cortés, H., González-Del Carmen, M., Leyva-Gómez, G., Rodríguez-Morales, M., Bustamante-Montes, L.P., Silva-Adaya, D., Pérez-Plasencia, C., Jacobo-Herrera, N., Reyes-Hernández, O.D., & Figueroa-González, G. (2023, April 20). Lithium as a Promising Anticancer Agent. In Encyclopedia. https://encyclopedia.pub/entry/43305
Villegas-Vázquez, Edgar Yebrán, et al. "Lithium as a Promising Anticancer Agent." Encyclopedia. Web. 20 April, 2023.
Lithium as a Promising Anticancer Agent
Edit

Lithium is a therapeutic cation used to treat bipolar disorders but also has some important features as an anti-cancer agent. Lithium formulations such as lithium acetoacetate (LiAcAc), lithium chloride (LiCl), lithium citrate (Li3C6H5O7), and lithium carbonate (Li2CO3) induce apoptosis, autophagy, and inhibition of tumor growth and also participate in the regulation of tumor proliferation, tumor invasion, and metastasis and cell cycle arrest. Moreover, lithium is synergistic with standard cancer therapies, enhancing their anti-tumor effects. In addition, lithium has a neuroprotective role in cancer patients, by improving their quality of life. Interestingly, nano-sized lithium enhances its anti-tumor activities and protects vital organs from the damage caused by lipid peroxidation during tumor development.

lithium cancer apoptosis autophagy anti-cancer effects

1. Introduction

Cancer is a disease characterized by the uncontrolled growth of certain cells. The phenotypic and genotypic complexity of cancer cells translates into the acquisition of functional capabilities, to continue to grow and spread. Capabilities for sustaining proliferative signaling, evading growth suppressors, non-mutational epigenetic reprogramming, avoiding immune destruction, enabling replicative immortality, tumor-promoting inflammation, polymorphic microbiomes, activating invasion and metastasis, inducing or accessing vasculature, senescent cells, genome instability and mutation, resisting cell death, deregulating cellular metabolism, and unlocking phenotypic plasticity are all hallmarks of cancer [1]. Metabolic alterations in cancer, known as the Warburg effect [2], have emerged as an alternative therapeutic pathway to deprive the body and tumor of glucose [3]. Therefore, there has been interest in ketogenic bodies (KBs), to evaluate the mechanism of glucose deprivation as a therapy in cancer. Among the KBs, acetoacetate (AcAc) is the most abundant and relevant [3] and, for in vitro and in vivo assays, AcAc is often used in the form of lithium acetoacetate (LiAcAc). Lithium has various therapeutic effects on neurotrophic factors, neurotransmitters, oxidative metabolism, apoptosis, neuronal structures, and glia [4][5].

2. The Effect of Lithium on Cancer Progression: Preclinical Approach

Lithium is a highly reactive alkali metal that exists as Li+ in seawater and in minerals [6]. The Li+ cation is therapeutically relevant, given that it has been used to treat bipolar disorders [7][8][9], Alzheimer’s disease [10][11], and as adjunctive therapy in thyroid cancer [12][13]. For bipolar mood disorders, achievable Li levels in plasma are around 0.5–1.0 mM (mEq/L) [14], whereas concentrations higher than 2 mM cause cytotoxicity in patients, resulting in nausea, fine tremors, diarrhea, confusion, slurred speech, and ataxia [14]. Lithium forms a complex with ATP and Mg2+, resulting in the bioactive form of lithium that modulates purine receptor activity in neuronal cells [15]. Lithium had been used over decades for bipolar disorder treatment, preventing relapses, and for the prophylaxis of manic episodes [16]. Some morphological abnormalities have been observed in patients with bipolar disorder. The neuroprotective activity of Li is related to the inhibition of its molecular targets, such as the enzymes inositol-monophosphate (IMPase), glycogen-synthase-kinase 3β (GSK3), and protein kinase C (PKC) [17]. Interestingly, these enzymes are deregulated in cancer, suggesting a potential role for Li, beyond bipolar disorders, in cancer treatment. Recent in vitro and in vivo studies reported the effects of lithium (lithium acetoacetate (LiAcAc), lithium chloride (LiCl), lithium citrate (Li3C6H5O7), and lithium carbonate (Li2CO3) on cell proliferation, tumor growth, and cancer metabolism. According to this evidence, LiAcAc inhibited cell proliferation in colon, breast, pancreatic, and neuroblastoma cancers [18].

3. Lithium Transport

The Li+ ion shares transport and permeation pathways with other ions, such as sodium [19]. Therefore, extracellular and intracellular concentrations of the Li+ ion depend on its ingestion [20]. However, LiAcAc requires specific transporters. In cells, the presence of monocarboxylate transporters (MCTs) is involved in the passive transport of lactate, pyruvate, D-β-hydroxybutyrate, and acetoacetate across the cell membrane [21]. MCT2 has the highest affinity constant (Km) for KBs such as D-β-hydroxybutyrate and acetoacetate [22], but these substrates also are transported by MCT1 [23] and MCT4, which have a lower affinity for them [24]. LiAcAc is imported into cells through MCT1 and MCT2, with Km values of 5.5 mM and 0.8 mM, respectively [22][24][25]. These values indicate that MCT2 has a higher affinity for LiAcAc compared to MCT1, therefore the cellular expression of MCT2 could determine the cellular response to Li+ ion exposure. Differential expression of MCTs in healthy and cancer tissues has been reported. In this way, MCT1 is expressed by healthy or cancer tissues [26]. Furthermore, MCT1 has a specific expression in cardiomyocytes, hepatocytes, and cells of the gastrointestinal tract (stomach, duodenum, jejunum, ileum, cecum, colon, and rectum) [25][27][28][29][30][31][32][33][34][35][36]. In addition, this transporter is found in erythrocytes, lymphocytes, and monocytes [27][37], in adipocytes [38][39] and in mammary glands [40]. Moreover, MCT1 is found in endothelial brain cells, astrocytes, oligodendrocytes, microglial cells, and the hypothalamus [41][42][43][44][45]. Furthermore, this transporter was found in the peripheral nervous system [46][47]. While MCT1 is a highly distributed transporter, MCT2 has a restricted distribution, being found in the liver and kidney, testis, and central nervous system [25][48][49][50][51]. The ratio of intracellular/extracellular lithium concentration is about five, suggesting a passive distribution of Li+ ion. The transport of lithium is in both directions across the cellular membrane through a Na+-dependent counter-transport system. Furthermore, Li+ ion enters the voltage-dependent Na+ channel, but the Na+-K+ pump mediates the lithium uptake but not its release in cells [52]. Therefore, neuronal cells maintained a ratio of internal-external Li+ ion below 1 mM in patients exposed to 1–2 mM of lithium [52].

Lithium Transporters in Cancer

Regarding MCTs, evidence indicates that MCT1 and MCT4 are expressed in cancer cells, whereas scant information is available regarding MCT2 and MCT3 expression in cancer. Expression of MCT1 was reported for lung (non-small cell lung carcinomas, NSCLC), stomach, colon, bladder, prostate, breast, ovarian cervical, head and neck cancers, and gliomas [53][54][55][56][57][58][59]. On the other hand, MCT4 is expressed in colon, bladder, and breast cancers, and gliomas [55][57][58][59]. In addition, while MCT2 is expressed in NSCLS, colon, breast, and ovarian cancers [55][59], MCT3 is found in NSCLC [59] (Figure 1).
Figure 1. Expression of monocarboxylate transporters (MCTs) in various cancers. MCTs are involved in the passive transport of lithium acetoacetate across cell membranes, and their abundance is associated with cancer. (-), no expression; (*), medium expression; (**), high expression; (***), very high expression level; ND, not data.
Furthermore, chaperone proteins of MCTs are important mediators in cancer. CD147 and CD44 function as chaperones of MCT1. In this way, the expression of MCT1 and CD147 correlated in bladder and ovarian carcinomas [59], whereas the expression of CD44 is associated with MCT1 expression in lung cancer [55].

4. Lithium as a Specific Inhibitor of GSK3β

The chlorinated form of lithium inhibits Glycogen Synthase Kinase 3β (GSK3β), which is a serine/threonine protein kinase. GSK3β phosphorylates β-catenin in the Wnt metabolic pathway, leading to growth arrest [60][61]. Moreover, GSK3β participates in cellular proliferation and survival by activating nuclear factor ϰB-dependent gene transcription, suggesting an active role of GSK3β in tumorigenesis in pancreatic, colorectal, and prostate cancers, and gliomas [62][63][64][65][66]. Since GSK3β is involved in repressing Wnt/β-catenin signaling, but also in maintaining cell survival and proliferation via the NF-ϰB pathway, the inhibition of this dual kinase activity by lithium might have a positive effect on cancer hallmarks.
The expression of GSK3β is higher in colon cancer cells and in colorectal cancer patients in comparison to normal counterparts. Furthermore, the inhibition of GSK3β activity resulted in apoptosis induction and reduced proliferation in cell culture [65]. Interestingly, lithium is capable of inhibiting GSK3β; consequently, lithium is a potential therapeutic agent. Inhibition of GSK3β by LiCl takes place through two mechanisms. One of them is a competitive mechanism between lithium and magnesium for binding to GSK3β [67][68]. The other mechanism involves the role of LiCl in the phosphorylation of the serine-9, located in the N-terminal, which is the main regulator region of GSK3β [69]. However, GSK3β has contradictory functions in cancer cells. On one hand, GSK3β phosphorylates p53, leading to its inhibition [70]. In this way, GSK3β binds to p53 in camptothecin-treated neuroblastoma cells, leading to increased mitochondrial apoptosis signaling and the expression of p21 and Bax [71][72].
Furthermore, when lithium blocks GSK3β activity, ovarian cancer cell proliferation was reduced and tumor suppressor activity was observed in nude mice inoculated with human ovarian cancer cells [73]. Moreover, proliferation was reduced in Li-treated human medullary thyroid TT cancer cells, due to the inactivation of GSK3β activity [74]

5. Effect of Lithium on Certain Cancer Hallmarks

5.1. Effect of Lithium on Apoptosis

The role of lithium as a proapoptotic or antiapoptotic compound is contradictory. In HepG2 cells, lithium confers resistance to the apoptosis induced by etoposide and camptothecin, by preventing the activation of caspase-8 and caspase-3 and by inhibiting the Bax translocation to mitochondria [75]. Therefore, lithium confers resistance to chemotherapy-induced apoptosis through GSK3β inhibition, disruption of GSK3β/p53 cooperation, and repression of CD95 expression [75].
On the other hand, in human SH-SY5Y neuroblastoma cells, lithium inhibits GSK3β, contributing to antiapoptotic signaling mechanisms [76]. Furthermore, LiCl induces the decrease of proapoptotic proteins p53 and Bax, while increasing Bcl-2 (a major antiapoptotic protein that controls mitochondrial membrane permeability) levels [77]. In addition, the effects of mitochondrial toxins might be attenuated by the inhibition of GSK3β, which can be achieved by the activation of the phosphatidylinositol 3-kinase (PI3K)/Akt pathway, which is inhibited by GSK3β through Akt-mediated phosphorylation of Ser-9 of GSK3β [78] or by GSK3β direct inhibitors such as lithium, suggesting that the inhibition of GSK3β provides protection against the activation of the apoptosis-associated cysteine protease caspase-3 [79].
In glioblastoma cells, low and higher LiCl doses had no effect on the expression of Bcl-2, procaspase-3, and PARP [80]. In contrast, in neuronal cells, Li blocks caspase-3 activation [81]. Therefore, cells of different origins (neuronal or glial) might have different results when treated with lithium. In cancer, several pathways might be dysregulated, and combinatory therapy with LiCl and pharmacological activators of the Notch1 pathway suppressed the hormonal secretion and reduces cell growth through apoptosis in medullary thyroid cancer (MTC) [82]. Lithium chloride enhances cell death induced by tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) in human prostate cancer cells [83].

5.2. Effect of Lithium on Autophagy

Lithium induces autophagy and enhances the clearance of aggregated proteins, such as mutant huntingtin and α-synucleins neuronal precursor cell lines and non-neuronal cells. Interestingly, this effect is not mediated by GSK3β, but it was mediated by inhibition of IMPase, leading to free inositol depletion and reducing myo-inositol-1,4,5-triphosphate (IP3) levels, resulting in a novel form of autophagy induction that is independent of the mammalian target rapamycin (mTOR) [84][85]. In addition, the inhibition of IMPase and GSK3β showed opposite effects on autophagy. On one hand, Li induced the inhibition of IMPase at lower doses (Ki = 0.8 mM), enhancing autophagy [85].
Additionally, lithium carbonate promotes autophagic vacuole formation in hepatocellular carcinoma (HCC) cells in vivo, suggesting that lithium-mediated autophagy might be a novel approach for treating hepatocellular carcinoma [86]. Interestingly, in HCC, combinatory treatment of lithium carbonate and rapamycin increases autolysosome formation and the expression of autophagy markers such as LC3β and LAMP1 [87]. Further, 5 mM lithium carbonate produced an antitumor effect, by arresting the cell cycle in G2/M-phase and increasing the number of apoptotic HCC cells, as well as inducing autophagy [88]. In corneal endothelial cells, treatment with lithium carbonate upregulated the expression of P62, Tmem74, Tm9sf1, and Tmem166, indicating that lithium increased autophagy that may have contributed to increased endothelial cell survival in a mouse model of Fuchs endothelial corneal dystrophy [89].

5.3. Effect of Lithium on Tumor Growth, Tumor Proliferation, Tumor Invasion and Metastasis, and Cell Cycle Arrest

Tumor cell growth is controlled by various signaling pathways, such as PI3K/Akt and the raf-1/mitogen-regulated extracellular kinase (MEK)/extracellular regulated kinase (ERK) pathway (raf-1/MEK/ERK). LiCl decreases hormonal secretion and suppresses the growth of MTC in vitro and in vivo due to GSK3β inhibition [74]. The raf-1 activation induces GSK3β phosphorylation, therefore the inactivation of GSK3β by LiCl might be a mechanism of rat-1-induced growth arrest [74].
Interestingly, low concentrations of LiCl (at 10, 20, 30, and 40 mM) promote the proliferation of mk3 and mk4 cells (metanephric mesenchyme cells), which play an essential role in nephron generation, by increasing the expression of the transcription factor Six2 (sine oculis homeobox homolog 2 gene). In contrast, higher concentrations of LiCl (50 mM) inhibit proliferation and downregulate the expression of Six2 [90]
Lithium causes nuclear localization of the claudin-1 and β-catenin that form the apical junctional complex, which is associated with colorectal cancer progression. In addition, lithium reduces colony formation, inhibiting cell migration and cell proliferation through G2/M cell cycle arrest. In colorectal cancer, the aberrant activation of epidermal growth factor (EGF) disrupts the association between E-cadherin and β-catenin, increasing cell proliferation and migration [1]. EGFR activates downstream kinases that modulate the GSK3β phosphorylation and consequently activate the Wnt/β-catenin pathway [91][92]. Since LiCl inhibited the tumorigenic effects of EGF, this cation might have tumor suppressor activity in colorectal cancer [93]. LiCl inhibits multiple-myeloma proliferation in a dose-dependent manner and triggers cell cycle arrest and apoptosis through inhibition of GSK3β, which is a crucial mediator of the Wnt/β-catenin pathway [94]. GSK3β regulates cadherin-11 at transcriptional and translational levels. Lithium inhibition of GSK3β suppresses cadherin-11, which is involved in cell–cell adhesion, cancer cell invasion, and metastasis of cancer [95]
In colon cancer, lithium reduces the expression of transforming growth factor-β-induced protein (TGFBIp) by inhibiting Smad3 phosphorylation through GSK3β inhibition [96]. In addition, Li inhibits cell migration of lymphatic endothelial cells and prevents metastasis by inhibiting TGFBIp-induced tumor lymphangiogenesis, resulting in the inhibition of colon cancer metastasis [96]. The effects of lithium on migration are greater than its effects on viability and occur at lower doses and earlier time points. Moreover, the migration effects of Li are reversible, suggesting that migration blockade is not due to cytotoxicity [97]

6. Anti-Inflammatory Activity of Lithium

In some types of cancer, before the cell becomes malignant, inflammatory conditions occur. Therefore, inflammation is associated with the proliferation and survival of cancer cells. In addition, this promotes angiogenesis and metastasis [98]. Since inflammation is associated with the development and progression of cancer, inflammatory biomarkers such as pro-inflammatory cytokines, tumor necrosis factor (TNF), interleukins 1 and 6, and chemokines can be used to monitor cancer progression [99]. In this way, the anti-inflammatory effect of lithium might be an alternative for prevention and monitoring of cancer. Furthermore, in animal models, LiCl treatment induced therapeutic effects on inflammation-mediated skeletal muscle wasting such as sepsis-induced muscle atrophy and cancer cachexia [100].
The anti-inflammatory activity of lithium results from its strong inhibitory effect on GSK3β, leading to a reduction in TNF-α production via attenuated activation of NF-κB and JNK signaling cascades [101] and induction of cytokine IL-10 [102]. The anti-inflammatory activity of lithium is due to the reduction of cyclooxygenase-2 expression, induction of the nitric oxide synthase expression, inhibition of interleukin 1β and TNFα, and increased production of IL-2 and IL-10 [103][104].

7. Synergism of Lithium with Standard Cancer Therapies

High concentrations of lithium (4–5 mM) inhibit inositol 1-monophosphatase, resulting in a reduction of phosphatidylinositol turnover. However, these high concentrations of Li+ ion have toxic effects in patients [14]. Reported serum Li+ ion levels for therapy were 0.8 to 1.2 mEq/L (0.8–1.2 mM) and the cytotoxicity effect began at 1.5 to 2 mEq/L (1.5–2 mM) [105]. Interestingly, 1 mM of LiCl had no effect on cellular metabolism or cell cycle but its combination with cisplatin or paclitaxel reduced metabolic activity in a serous ovarian cancer cell line (OVCA 433) or primary cultures. However, LiCl by itself or in combination with cisplatin or paclitaxel had no significant effect on cellular proliferation, suggesting that the effects of LiCl are mainly related to cellular metabolism rather than proliferation [105].
Conventional cancer therapies such as chemotherapy and radiotherapy inhibit cell division, causing neurocognitive deterioration. Preliminary studies have shown that lithium has a neuroprotective role in cancer patients undergoing radiotherapy or chemotherapy treatment, improving their quality of life [106]. Combination of LiCl with a low dose or IC50 dose of etoposide enhances apoptosis and reduces the percentage of cells in the G2/M phase in prostate cancer LNCap cells [107]. In pancreatic ductal adenocarcinoma cells, lithium increases the anti-tumoral efficacy of gemcitabine [108]. Additionally, histone deacetylase inhibitors with lithium resulted in an effective combinatory therapy, due to inducing the Notch1 pathway and inhibiting GSK3β activity, respectively. [82]
Chemotherapy-induced peripheral neuropathy (CIPN) is an adverse effect of cancer treatment using taxanes, such as paclitaxel. Adding lithium to taxane treatment prevents calcium overloading in mitochondria, resulting in the prevention of calpain activation and preventing the loss of cell functions related to the progression of peripheral neuropathy [109], suggesting that lithium could be used to prevent the adverse effects of cancer treatment with only taxane administration [110].

8. Contradictory Effects of Lithium

Prolonged treatment with lithium causes renal, endocrine, and dermatological side effects. Four weeks of therapy with lithium caused renal tubular concentration defects, polyuria, polydipsia, and nephrogenic diabetes insipidus [111]. Administration of Li2CO3 induced a cutaneous adverse reaction [112] and parathyroid dysfunction [113]. Furthermore, lithium treatment caused a higher prevalence of hypercalcemia in a cohort of bipolar patients [114]. In addition, there is a possibility of teratogenicity activity with lithium treatment [115].
The inhibition of GSK3β using lithium has contradictory effects. On one hand, chemoresistance to apoptosis is observed, but on the other hand, it induces tumor suppressor activity and reduces proliferation. Those effects depend on the type of cancer, the origin of cells, and doses. The anti-cancer effect of lithium has been reported for colorectal cancer, neuroblastoma cells, ovarian cancer cells, medullary thyroid TT cancer cells, esophageal cancer, medulloblastoma, and glioblastoma multiforme, hematological tumors (multiple myeloma), breast cancer cells, MTC, prostate cancer cells, pancreatic ductal adenocarcinoma cells, corneal endothelial cells, and colon cancer cells. However, opposite effects of lithium have been reported for neuroblastoma, hepatoblastoma cells, and malignant glioma cell lines.
Lithium induces apoptosis in promyelocytic leukemia cells, but a small fraction of these cells remain viable and survive the cytotoxic effect of this cation, resulting in altered gene expression patterns [116], suggesting cell tolerance to lithium in culture [117]. Resistance to lithium is a consequence of its activity. Lithium appears to induce apoptosis by increasing the bax/bcl-2 ratio (bax: pro-apoptotic gene and bcl-2: anti-apoptotic gene). Lithium resistance then results when this ratio is reversed, leading to lithium resistance accompanied by DNA fragmentation, and decreasing cell viability [118].

References

  1. Hanahan, D. Hallmarks of cancer: New dimensions. Cancer Discov. 2022, 12, 31–46.
  2. Warburg, O. The Metabolism of Tumors; Richard R. Smith. Inc.: New York, NY, USA, 1931.
  3. Poff, A.; Koutnik, A.P.; Egan, K.M.; Sahebjam, S.; D’Agostino, D.; Kumar, N.B. Targeting the Warburg effect for cancer treatment: Ketogenic diets for management of glioma. In Seminars in Cancer Biology; Elsevier: Amsterdam, The Netherlands, 2019.
  4. Won, E.; Kim, Y.-K. An oldie but goodie: Lithium in the treatment of bipolar disorder through neuroprotective and neurotrophic mechanisms. Int. J. Mol. Sci. 2017, 18, 2679.
  5. Elbe, D. Stahl’s Essential Psychopharmacology: Neuroscientific Basis and Practical Applications. J. Can. Acad. Child Adolesc. Psychiatry 2010, 19, 230.
  6. de Freitas, D.M.; Leverson, B.D.; Goossens, J.L. Lithium in medicine: Mechanisms of action. In The Alkali Metal Ions: Their Role for Life; Springer: Berlin/Heidelberg, Germany, 2016; pp. 557–584.
  7. Edition, F. Diagnostic and statistical manual of mental disorders. Am. Psychiatr. Assoc. 2013, 21, 591–643.
  8. Walker, E.R.; McGee, R.E.; Druss, B.G. Mortality in mental disorders and global disease burden implications: A systematic review and meta-analysis. JAMA Psychiatry 2015, 72, 334–341.
  9. Wong, M.M. Management of bipolar II disorder. Indian J. Psychol. Med. 2011, 33, 18–28.
  10. Kessing, L.V.; Søndergård, L.; Forman, J.L.; Andersen, P.K. Lithium treatment and risk of dementia. Arch. Gen. Psychiatry 2008, 65, 1331–1335.
  11. Nitrini, R.; Caramelli, P.; Herrera, E.; Bahia, V.S.; Caixeta, L.F.; Radanovic, M.; Anghinah, R.; Charchat-Fichman, H.; Porto, C.S.; Carthery, M.T.; et al. Incidence of dementia in a community-dwelling Brazilian population. Alzheimer Dis. Assoc. Disord. 2004, 18, 241–246.
  12. Köhrle, J. Local activation and inactivation of thyroid hormones: The deiodinase family. Mol. Cell. Endocrinol. 1999, 151, 103–119.
  13. Berens, S.C.; Bernstein, R.S.; Robbins, J.; Wolff, J. Antithyroid effects of lithium. J. Clin. Investig. 1970, 49, 1357–1367.
  14. Kurita, M.; Mashiko, H.; Rai, M.; Kumasaka, T.; Kouno, S.-I.; Niwa, S.-I.; Nakahata, N. Lithium chloride at a therapeutic concentration reduces Ca2+ response in protein kinase C down-regulated human astrocytoma cells. Eur. J. Pharmacol. 2002, 442, 17–22.
  15. Briggs, K.T.; Giulian, G.G.; Li, G.; Kao, J.P.; Marino, J.P. A A Molecular Model for Lithium’s Bioactive Form. Biophys. J. 2016, 111, 294–300.
  16. Volkmann, C.; Bschor, T.; Köhler, S. Lithium treatment over the lifespan in bipolar disorders. Front. Psychiatry 2020, 11, 377.
  17. Ochoa, E.L.M. Lithium as a neuroprotective agent for bipolar disorder: An overview. Cell. Mol. Neurobiol. 2022, 42, 85–97.
  18. Vidali, S.; Aminzadeh-Gohari, S.; Vatrinet, R.; Iommarini, L.; Porcelli, A.M.; Kofler, B.; Feichtinger, R.G. Lithium and not acetoacetate influences the growth of cells treated with lithium acetoacetate. Int. J. Mol. Sci. 2019, 20, 3104.
  19. Jakobsson, E.; Argüello-Miranda, O.; Chiu, S.-W.; Fazal, Z.; Kruczek, J.; Nunez-Corrales, S.; Pandit, S.; Pritchet, L. Towards a unified understanding of lithium action in basic biology and its significance for applied biology. J. Membr. Biol. 2017, 250, 587–604.
  20. de Roos, N.M.; de Vries, J.H.; Katan, M.B. Serum lithium as a compliance marker for food and supplement intake. Am. J. Clin. Nutr. 2001, 73, 75–79.
  21. Pérez-Escuredo, J.; Van Hée, V.F.; Sboarina, M.; Falces, J.; Payen, V.L.; Pellerin, L.; Sonveaux, P. Monocarboxylate transporters in the brain and in cancer. Biochim. Biophys. Acta Mol. Cell Res. 2016, 1863, 2481–2497.
  22. Bröer, S.; Bröer, A.; Schneider, H.-P.; Stegen, C.; Halestrap, A.P.; Deitmer, J.W. Characterization of the high-affinity monocarboxylate transporter MCT2 in Xenopus laevis oocytes. Biochem. J. 1999, 341, 529–535.
  23. Bröer, S.; Schneider, H.-P.; Bröer, A.; Rahman, B.; Hamprecht, B.; Deitmer, J.W. Characterization of the monocarboxylate transporter 1 expressed in Xenopus laevis oocytes by changes in cytosolic pH. Biochem. J. 1998, 333, 167–174.
  24. Halestrap, A.P. The monocarboxylate transporter family—Structure and functional characterization. IUBMB Life 2012, 64, 1–9.
  25. Garcia, C.K.; Brown, M.S.; Pathak, R.K.; Goldstein, J.L. cDNA Cloning of MCT2, a Second Monocarboxylate Transporter Expressed in Different Cells than MCT1. J. Biol. Chem. 1995, 270, 1843–1849.
  26. Halestrap, A.P.; Meredith, D. The SLC16 gene family—From monocarboxylate transporters (MCTs) to aromatic amino acid transporters and beyond. Pflügers Arch. 2004, 447, 619–628.
  27. Garcia, C.K.; Goldstein, J.L.; Pathak, R.K.; Anderson, R.G.; Brown, M.S. Molecular characterization of a membrane transporter for lactate, pyruvate, and other monocarboxylates: Implications for the Cori cycle. Cell 1994, 76, 865–873.
  28. Jóhannsson, E.; Nagelhus, E.A.; Mccullagh, K.J.; Sejersted, O.M.; Blackstad, T.W.; Bonen, A.; Ottersen, O.P. Cellular and subcellular expression of the monocarboxylate transporter MCT1 in rat heart: A high-resolution immunogold analysis. Circ. Res. 1997, 80, 400–407.
  29. Kirat, D.; Inoue, H.; Iwano, H.; Yokota, H.; Taniyama, H.; Kato, S. Monocarboxylate transporter 1 (MCT1) in the liver of pre-ruminant and adult bovines. Vet. J. 2007, 173, 124–130.
  30. Tamai, I.; Takanaga, H.; Ogihara, T.; Higashida, H.; Maeda, H.; Sai, Y.; Tsuji, A. Participation of a proton-cotransporter, MCT1, in the intestinal transport of monocarboxylic acids. Biochem. Biophys. Res. Commun. 1995, 214, 482–489.
  31. Ritzhaupt, A.; Wood, I.S.; Ellis, A.; Hosie, K.B.; Shirazi-Beechey, S.P. Identification and characterization of a monocarboxylate transporter (MCT1) in pig and human colon: Its potential to transport l-lactate as well as butyrate. J. Physiol. 1998, 513, 719–732.
  32. Orsenigo, M.N.; Tosco, M.; Bazzini, C.; Laforenza, U.; Faelli, A. A monocarboxylate transporter MCT1 is located at the basolateral pole of rat jejunum. Exp. Physiol. 1999, 84, 1033–1042.
  33. Kirat, D.; Inoue, H.; Iwano, H.; Hirayama, K.; Yokota, H.; Taniyama, H.; Kato, S. Monocarboxylate transporter 1 gene expression in the ovine gastrointestinal tract. Vet. J. 2006, 171, 462–467.
  34. Iwanaga, T.; Takebe, K.; Kato, I.; Karaki, S.-I.; Kuwahara, A. Cellular expression of monocarboxylate transporters (MCT) in the digestive tract of the mouse, rat, and humans, with special reference to slc5a8. Biomed. Res. 2006, 27, 243–254.
  35. Shimoyama, Y.; Kirat, D.; Akihara, Y.; Kawasako, K.; Komine, M.; Hirayama, K.; Matsuda, K.; Okamoto, M.; Iwano, H.; Kato, S.; et al. Expression of monocarboxylate transporter 1 (MCT1) in the dog intestine. J. Vet. Med. Sci. 2007, 69, 599–604.
  36. Welter, H.; Claus, R. Expression of the monocarboxylate transporter 1 (MCT1) in cells of the porcine intestine. Cell Biol. Int. 2008, 32, 638–645.
  37. Poole, R.C.; Halestrap, A.P. N-terminal protein sequence analysis of the rabbit erythrocyte lactate transporter suggests identity with the cloned monocarboxylate transport protein MCT1. Biochem. J. 1994, 303, 755–759.
  38. de Heredia, F.P.; Wood, I.S.; Trayhurn, P. Hypoxia stimulates lactate release and modulates monocarboxylate transporter (MCT1, MCT2, and MCT4) expression in human adipocytes. Pflügers Arch.-Eur. J. Physiol. 2010, 459, 509–518.
  39. Hajduch, E.; Heyes, R.R.; Watt, P.W.; Hundal, H.S. Lactate transport in rat adipocytes: Identification of monocarboxylate transporter 1 (MCT1) and its modulation during streptozotocin-induced diabetes. FEBS Lett. 2000, 479, 89–92.
  40. Takebe, K.; Nio-Kobayashi, J.; Takahashi-Iwanaga, H.; Yajima, T.; Iwanaga, T. Cellular expression of a monocarboxylate transporter (MCT1) in the mammary gland and sebaceous gland of mice. Histochem. Cell Biol. 2009, 131, 401–409.
  41. Mac, M.; Nałęcz, K.A. Expression of monocarboxylic acid transporters (MCT) in brain cells: Implication for branched chain α-ketoacids transport in neurons. Neurochem. Int. 2003, 43, 305–309.
  42. Hanu, R.; McKenna, M.; O’Neill, A.; Resneck, W.G.; Bloch, R.J. Monocarboxylic acid transporters, MCT1 and MCT2, in cortical astrocytes in vitro and in vivo. Am. J. Physiol. Physiol. 2000, 278, C921–C930.
  43. Lee, Y.; Morrison, B.M.; Li, Y.; Lengacher, S.; Farah, M.H.; Hoffman, P.N.; Liu, Y.; Tsingalia, A.; Jin, L.; Zhang, P.-W.; et al. Oligodendroglia metabolically support axons and contribute to neurodegeneration. Nature 2012, 487, 443–448.
  44. Moreira, T.J.; Pierre, K.; Maekawa, F.; Repond, C.; Cebere, A.; Liljequist, S.; Pellerin, L. Enhanced cerebral expression of MCT1 and MCT2 in a rat ischemia model occurs in activated microglial cells. J. Cereb. Blood Flow Metab. 2009, 29, 1273–1283.
  45. Ainscow, E.K.; Mirshamsi, S.; Tang, T.; Ashford, M.L.; Rutter, G.A. Dynamic imaging of free cytosolic ATP concentration during fuel sensing by rat hypothalamic neurones: Evidence for ATP-independent control of ATP-sensitive K+ channels. J. Physiol. 2002, 544, 429–445.
  46. Domènech-Estévez, E.; Baloui, H.; Repond, C.; Rosafio, K.; Médard, J.-J.; Tricaud, N.; Pellerin, L.; Chrast, R. Distribution of monocarboxylate transporters in the peripheral nervous system suggests putative roles in lactate shuttling and myelination. J. Neurosci. 2015, 35, 4151–4156.
  47. Morrison, B.M.; Tsingalia, A.; Vidensky, S.; Lee, Y.; Jin, L.; Farah, M.H.; Lengacher, S.; Magistretti, P.J.; Pellerin, L.; Rothstein, J.D. Deficiency in monocarboxylate transporter 1 (MCT1) in mice delays regeneration of peripheral nerves following sciatic nerve crush. Exp. Neurol. 2015, 263, 325–338.
  48. Boussouar, F.; Mauduit, C.; Tabone, E.; Pellerin, L.; Magistretti, P.J.; Benahmed, M. Developmental and hormonal regulation of the monocarboxylate transporter 2 (MCT2) expression in the mouse germ cells. Biol. Reprod. 2003, 69, 1069–1078.
  49. Pierre, K.; Pellerin, L.; Debernardi, R.; Riederer, B.; Magistretti, P.J. Cell-specific localization of monocarboxylate transporters, MCT1 and MCT2, in the adult mouse brain revealed by double immunohistochemical labeling and confocal microscopy. Neuroscience 2000, 100, 617–627.
  50. Pellerin, L.; Halestrap, A.P.; Pierre, K. Cellular and subcellular distribution of monocarboxylate transporters in cultured brain cells and in the adult brain. J. Neurosci. Res. 2005, 79, 55–64.
  51. Pierre, K.; Magistretti, P.J.; Pellerin, L. MCT2 is a major neuronal monocarboxylate transporter in the adult mouse brain. J. Cereb. Blood Flow Metab. 2002, 22, 586–595.
  52. Reiser, G.; Duhm, J. Transport pathways for lithium ions in neuroblastoma× glioma hybrid cells at ‘therapeutic’concentrations of Li+. Brain Res. 1982, 252, 247–258.
  53. Sonveaux, P.; Vegran, F.; Schroeder, T.; Wergin, M.C.; Verrax, J.; Rabbani, Z.N.; De Saedeleer, C.J.; Kennedy, K.M.; Diepart, C.; Jordan, B.F.; et al. Targeting lactate-fueled respiration selectively kills hypoxic tumor cells in mice. J. Clin. Investig. 2008, 118, 3930–3942.
  54. Baltazar, F.; Pinheiro, C.; Morais-Santos, F.; Azevedo-Silva, J.; Queiros, O.; Preto, A.; Casal, M. Monocarboxylate transporters as targets and mediators in cancer therapy response. Histol. Histopathol. 2014, 29, 1511–1524.
  55. Pinheiro, C.; Reis, R.M.; Ricardo, S.; Longatto-Filho, A.; Schmitt, F.; Baltazar, F. Expression of monocarboxylate transporters 1, 2, and 4 in human tumours and their association with CD147 and CD44. J. Biomed. Biotechnol. 2010, 2010, 427694.
  56. Kennedy, K.M.; Dewhirst, M.W. Tumor metabolism of lactate: The influence and therapeutic potential for MCT and CD147 regulation. Future Oncol. 2010, 6, 127–148.
  57. Pinheiro, C.; Longatto-Filho, A.; Azevedo-Silva, J.; Casal, M.; Schmitt, F.C.; Baltazar, F. Role of monocarboxylate transporters in human cancers: State of the art. J. Bioenerg. Biomembr. 2012, 44, 127–139.
  58. Miranda-Gonçalves, V.; Honavar, M.; Pinheiro, C.; Martinho, O.; Pires, M.M.; Pinheiro, C.; Cordeiro, M.; Bebiano, G.; Costa, P.; Palmeirim, I.; et al. Monocarboxylate transporters (MCTs) in gliomas: Expression and exploitation as therapeutic targets. Neuro-Oncol. 2013, 15, 172–188.
  59. Afonso, J.; Santos, L.L.; Miranda-Gonçalves, V.; Morais, A.; Amaro, T.; Longatto-Filho, A.; Baltazar, F. CD147 and MCT1-potential partners in bladder cancer aggressiveness and cisplatin resistance. Mol. Carcinog. 2015, 54, 1451–1466.
  60. Wang, Z.; Smith, K.S.; Murphy, M.; Piloto, O.; Somervaille, T.C.P.; Cleary, M.L. Glycogen synthase kinase 3 in MLL leukaemia maintenance and targeted therapy. Nature 2008, 455, 1205–1209.
  61. Woodgett, J.R. Regulation and functions of the glycogen synthase kinase-3 subfamily. Semin. Cancer Biol. 1994, 5, 269–275.
  62. Ougolkov, A.V.; Fernandez-Zapico, M.E.; Savoy, D.N.; Urrutia, R.A.; Billadeau, D.D. Glycogen synthase kinase-3β participates in nuclear factor κB–mediated gene transcription and cell survival in pancreatic cancer cells. Cancer Res. 2005, 65, 2076–2081.
  63. Kotliarova, S.; Pastorino, S.; Kovell, L.C.; Kotliarov, Y.; Song, H.; Zhang, W.; Bailey, R.; Maric, D.; Zenklusen, J.C.; Lee, J.; et al. Glycogen synthase kinase-3 inhibition induces glioma cell death through c-MYC, nuclear factor-κB, and glucose regulation. Cancer Res. 2008, 68, 6643–6651.
  64. Marchand, B.; Tremblay, I.; Cagnol, S.; Boucher, M.-J. Inhibition of glycogen synthase kinase-3 activity triggers an apoptotic response in pancreatic cancer cells through JNK-dependent mechanisms. Carcinogenesis 2012, 33, 529–537.
  65. Shakoori, A.; Ougolkov, A.; Yu, Z.W.; Zhang, B.; Modarressi, M.H.; Billadeau, D.D.; Mai, M.; Takahashi, Y.; Minamoto, T. Deregulated GSK3β activity in colorectal cancer: Its association with tumor cell survival and proliferation. Biochem. Biophys. Res. Commun. 2005, 334, 1365–1373.
  66. Mazor, M.; Kawano, Y.; Zhu, H.; Waxman, J.; Kypta, R.M. Inhibition of glycogen synthase kinase-3 represses androgen receptor activity and prostate cancer cell growth. Oncogene 2004, 23, 7882–7892.
  67. Ryves, W.; Harwood, A.J. Lithium inhibits glycogen synthase kinase-3 by competition for magnesium. Biochem. Biophys. Res. Commun. 2001, 280, 720–725.
  68. Klein, P.S.; Melton, D.A. A molecular mechanism for the effect of lithium on development. Proc. Natl. Acad. Sci. USA 1996, 93, 8455–8459.
  69. Li, X.; Friedman, A.B.; Zhu, W.; Wang, L.; Boswell, S.; May, R.S.; Davis, L.L.; Jope, R.S. Lithium regulates glycogen synthase kinase-3β in human peripheral blood mononuclear cells: Implication in the treatment of bipolar disorder. Biol. Psychiatry 2007, 61, 216–222.
  70. Qu, L.; Huang, S.; Baltzis, D.; Rivas-Estilla, A.-M.; Pluquet, O.; Hatzoglou, M.; Koumenis, C.; Taya, Y.; Yoshimura, A.; Koromilas, A.E. Endoplasmic reticulum stress induces p53 cytoplasmic localization and prevents p53-dependent apoptosis by a pathway involving glycogen synthase kinase-3β. Genes Dev. 2004, 18, 261–277.
  71. Watcharasit, P.; Bijur, G.N.; Song, L.; Zhu, J.; Chen, X.; Jope, R.S. Glycogen synthase kinase-3β (GSK3β) binds to and promotes the actions of p53. J. Biol. Chem. 2003, 278, 48872–48879.
  72. Watcharasit, P.; Bijur, G.N.; Zmijewski, J.W.; Song, L.; Zmijewska, A.; Chen, X.; Johnson, G.V.W.; Jope, R.S. Direct, activating interaction between glycogen synthase kinase-3β and p53 after DNA damage. Proc. Natl. Acad. Sci. USA 2002, 99, 7951–7955.
  73. Cao, Q.; Lu, X.; Feng, Y.-J. Glycogen synthase kinase-3β positively regulates the proliferation of human ovarian cancer cells. Cell Res. 2006, 16, 671–677.
  74. Kunnimalaiyaan, M.; Vaccaro, A.M.; Ndiaye, M.A.; Chen, H. Inactivation of glycogen synthase kinase-3β, a downstream target of the raf-1 pathway, is associated with growth suppression in medullary thyroid cancer cells. Mol. Cancer Ther. 2007, 6, 1151–1158.
  75. Beurel, E.; Kornprobst, M.; Eggelpoël, M.-J.B.-V.; Ruiz-Ruiz, C.; Cadoret, A.; Capeau, J.; Desbois-Mouthon, C. GSK-3β inhibition by lithium confers resistance to chemotherapy-induced apoptosis through the repression of CD95 (Fas/APO-1) expression. Exp. Cell Res. 2004, 300, 354–364.
  76. Bijur, G.N.; De Sarno, P.; Jope, R.S. Glycogen synthase kinase-3β facilitates staurosporine-and heat shock-induced apoptosis: Protection by lithium. J. Biol. Chem. 2000, 275, 7583–7590.
  77. Chen, R.-W.; Chuang, D.-M. Long term lithium treatment suppresses p53 and Bax expression but increases Bcl-2 expression: A prominent role in neuroprotection against excitotoxicity. J. Biol. Chem. 1999, 274, 6039–6042.
  78. Cross, D.A.; Alessi, D.R.; Cohen, P.; Andjelkovich, M.; Hemmings, B.A. Inhibition of glycogen synthase kinase-3 by insulin mediated by protein kinase B. Nature 1995, 378, 785–789.
  79. King, T.D.; Bijur, G.N.; Jope, R.S. Caspase-3 activation induced by inhibition of mitochondrial complex I is facilitated by glycogen synthase kinase-3β and attenuated by lithium. Brain Res. 2001, 919, 106–114.
  80. Karlovic, D.; Jakopec, S.; Dubravcic, K.; Batinic, D.; Buljan, D.; Osmak, M. Lithium increases expression of p21WAF/Cip1 and survivin in human glioblastoma cells. Cell Biol. Toxicol. 2007, 23, 83–90.
  81. Mora, A.; Sabio, G.; Alonso, J.C.; Soler, G.; Centeno, F. Different dependence of lithium and valproate on PI3K/PKB pathway. Bipolar Disord. 2002, 4, 195–200.
  82. Adler, J.T.; Hottinger, D.G.; Kunnimalaiyaan, M.; Chen, H. Inhibition of growth in medullary thyroid cancer cells with histone deacetylase inhibitors and lithium chloride. J. Surg. Res. 2010, 159, 640–644.
  83. Liao, X.; Zhang, L.; Thrasher, J.B.; Du, J.; Li, B. Glycogen synthase kinase-3β suppression eliminates tumor necrosis factor-related apoptosis-inducing ligand resistance in prostate cancer. Mol. Cancer Ther. 2003, 2, 1215–1222.
  84. Sarkar, S.; Rubinsztein, D.C. Inositol and IP3 levels regulate autophagy—Biology and therapeutic speculations. Autophagy 2006, 2, 132–134.
  85. Sarkar, S.; Floto, R.A.; Berger, Z.; Imarisio, S.; Cordenier, A.; Pasco, M.; Cook, L.J.; Rubinsztein, D.C. Lithium induces autophagy by inhibiting inositol monophosphatase. J. Cell Biol. 2005, 170, 1101–1111.
  86. Taskaeva, I.; Bgatova, N. Ultrastructural and immunofluorescent analysis of lithium effects on autophagy in hepatocellular carcinoma cells. Asian Pac. J. Cancer Biol. 2018, 3, 83–87.
  87. Taskaeva, I.S.; Bgatova, N.P.; Solovieva, A.O. Autophagy in Hepatocellular Carcinoma-29 after Single or Combined Administration of Lithium Carbonate and Rapamycin. Cell Tissue Biol. 2019, 13, 353–359.
  88. Taskaeva, Y.S.; Bgatova, N.P.; Dossymbekova, R.S.; Solovieva, A.O.; Miroshnichenko, S.M.; Sharipov, K.O.; Tungushbaeva, Z.B. In vitro effects of lithium carbonate on cell cycle, apoptosis, and autophagy in hepatocellular carcinoma-29 cells. Bull. Exp. Biol. Med. 2020, 170, 246–250.
  89. Kim, E.C.; Meng, H.; Jun, A.S. Lithium treatment increases endothelial cell survival and autophagy in a mouse model of Fuchs endothelial corneal dystrophy. Br. J. Ophthalmol. 2013, 97, 1068–1073.
  90. Liu, J.; Ju, P.; Zhou, Y.; Zhao, Y.; Xie, Y.; Long, Y.; Gu, Y.; Ni, D.; Lyv, Z.; Mao, Z.; et al. Six2 is a coordinator of LiCl-induced cell proliferation and apoptosis. Int. J. Mol. Sci. 2016, 17, 1504.
  91. Grimes, C.A.; Jope, R.S. The multifaceted roles of glycogen synthase kinase 3β in cellular signaling. Prog. Neurobiol. 2001, 65, 391–426.
  92. Ding, Q.; Xia, W.; Liu, J.-C.; Yang, J.-Y.; Lee, D.-F.; Xia, J.; Bartholomeusz, G.; Li, Y.; Pan, Y.; Li, Z.; et al. associates with and primes GSK-3β for its inactivation resulting in upregulation of β-catenin. Mol. Cell 2005, 19, 159–170.
  93. Díaz, M.; Vidal, F.; Cruz, A.L.; De Araujo, W.M.; Tanaka, M.N.; Viola, J.P.; Morgado-Diaz, J.A. Lithium reduces tumorigenic potential in response to EGF signaling in human colorectal cancer cells. Int. J. Oncol. 2011, 38, 1365–1373.
  94. Yao, R.; Sun, X.; Xie, Y.; Liu, L.; Han, D.; Yao, Y.; Li, H.; Li, Z.; Xu, K. Lithium chloride inhibits cell survival, overcomes drug resistance, and triggers apoptosis in multiple myeloma via activation of the Wnt/β-catenin pathway. Am. J. Transl. Res. 2018, 10, 2610.
  95. Farina, A.K.; Bong, Y.-S.; Feltes, C.M.; Byers, S.W. Post-transcriptional regulation of cadherin-11 expression by GSK-3 and β-catenin in prostate and breast cancer cells. PLoS ONE 2009, 4, e4797.
  96. Maeng, Y.-S.; Lee, R.; Lee, B.; Choi, S.-I.; Kim, E.K. Lithium inhibits tumor lymphangiogenesis and metastasis through the inhibition of TGFBIp expression in cancer cells. Sci. Rep. 2016, 6, 20739.
  97. Nowicki, M.O.; Dmitrieva, N.; Stein, A.M.; Cutter, J.L.; Godlewski, J.; Saeki, Y.; Nita, M.; Berens, M.E.; Sander, L.M.; Newton, H.B.; et al. Lithium inhibits invasion of glioma cells; possible involvement of glycogen synthase kinase-3. Neuro-Oncology 2008, 10, 690–699.
  98. Mantovani, A.; Allavena, P.; Sica, A.; Balkwill, F. Cancer-related inflammation. Nature 2008, 454, 436–444.
  99. Singh, N.; Baby, D.; Rajguru, J.P.; Patil, P.B.; Thakkannavar, S.S.; Pujari, V.B. Inflammation and cancer. Ann. Afr. Med. 2019, 18, 121.
  100. Lee, J.-H.; Kim, S.-W.; Kim, J.-H.; Kim, H.-J.; Um, J.; Jung, D.-W.; Williams, D. Lithium chloride protects against sepsis-induced skeletal muscle atrophy and cancer cachexia. Cells 2021, 10, 1017.
  101. Wang, M.J.; Huang, H.Y.; Chen, W.F.; Chang, H.F.; Kuo, J.S. Glycogen synthase kinase-3β inactivation inhibits tumor necrosis factor-α production in microglia by modulating nuclear factor κB and MLK3/JNK signaling cascades. J. Neuroinflamm. 2010, 7, 99.
  102. Huang, W.-C.; Lin, Y.-S.; Wang, C.-Y.; Tsai, C.-C.; Tseng, H.-C.; Chen, C.-L.; Lu, P.-J.; Chen, P.-S.; Qian, L.; Hong, J.-S.; et al. Glycogen synthase kinase-3 negatively regulates anti-inflammatory interleukin-10 for lipopolysaccharide-induced iNOS/NO biosynthesis and RANTES production in microglial cells. Immunology 2009, 128, e275–e286.
  103. Nahman, S.; Belmaker, R.; Azab, A.N. Effects of lithium on lipopolysaccharide-induced inflammation in rat primary glia cells. Endotoxin Res. 2012, 18, 447–458.
  104. Nassar, A.; Azab, A.N. Effects of lithium on inflammation. ACS Chem. Neurosci. 2014, 5, 451–458.
  105. Novetsky, A.P.; Thompson, D.M.; Zighelboim, I.; Thaker, P.H.; Powell, M.A.; Mutch, D.G.; Goodfellow, P.J. Lithium chloride and inhibition of glycogen synthase kinase 3β as a potential therapy for serous ovarian cancer. Int. J. Gynecol. Cancer 2013, 23, 361–366.
  106. Khasraw, M.; Ashley, D.; Wheeler, G.; Berk, M. Using lithium as a neuroprotective agent in patients with cancer. BMC Med. 2012, 10, 131.
  107. Hossein, G.; Janzamin, E.; Azimian-Zavareh, V. Effect of lithium chloride and antineoplastic drugs on survival and cell cycle of androgen-dependent prostate cancer LNCap cells. Indian J. Pharmacol. 2012, 44, 714.
  108. Peng, Z.; Ji, Z.; Mei, F.; Lu, M.; Ou, Y.; Cheng, X. Lithium inhibits tumorigenic potential of PDA cells through targeting hedgehog-GLI signaling pathway. PLoS ONE 2013, 8, e61457.
  109. Boehmerle, W.; Zhang, K.; Sivula, M.; Heidrich, F.M.; Lee, Y.; Jordt, S.-E.; Ehrlich, B.E. Chronic exposure to paclitaxel diminishes phosphoinositide signaling by calpain-mediated neuronal calcium sensor-1 degradation. Proc. Natl. Acad. Sci. USA 2007, 104, 11103–11108.
  110. Ibrahim, E.Y.; Ehrlich, B.E. Prevention of chemotherapy-induced peripheral neuropathy: A review of recent findings. Crit. Rev. Oncol./Hematol. 2020, 145, 102831.
  111. Walker, R.J.; Weggery, S.; Bedford, J.J.; Mcdonald, F.J.; Ellis, G.; Leader, J.P. Lithium-induced reduction in urinary concentrating ability and urinary aquaporin 2 (AQP2) excretion in healthy volunteers. Kidney Int. 2005, 67, 291–294.
  112. Pinna, M.; Manchia, M.; Puddu, S.; Minnai, G.; Tondo, L.; Salis, P. Cutaneous adverse reaction during lithium treatment: A case report and updated systematic review with meta-analysis. Int. J. Bipolar Disord. 2017, 5, 20.
  113. Nair, C.G.; Menon, R.; Jacob, P.; Babu, M. Lithium-induced parathyroid dysfunction: A new case. Indian J. Endocrinol. Metab. 2013, 17, 930–932.
  114. Meehan, A.D.; Udumyan, R.; Kardell, M.; Landén, M.; Järhult, J.; Wallin, G. Lithium-associated hypercalcemia: Pathophysiology, prevalence, management. World J. Surg. 2018, 42, 415–424.
  115. Poels, E.M.P.; Bijma, H.H.; Galbally, M.; Bergink, V. Lithium during pregnancy and after delivery: A review. Int. J. Bipolar Disord. 2018, 6, 26.
  116. Matsebatlela, T.M.; Mogodiri, R.K.; Hart, D.A.; Gallicchio, V.S.; Becker, R.W. Resistance to lithium-induced apoptosis in a lithium tolerant clone of HL-60 promyelocytes. J. Trace Microprobe Tech. 2000, 18, 163–170.
  117. Lucas, K.C.; Hart, D.A.; Becker, R.W. Porcine proximal tubular cells (LLC-PK1) are able to tolerate high levels of lithium chloride in vitro: Assessment of the influence of 1–20 mM LiCl on cell death and alterations in cell biology and biochemistry. Cell Biol. Int. 2010, 34, 225–233.
  118. Matsebatlela, T.; Gallicchio, V.; Becker, R. Lithium modulates cancer cell growth, apoptosis, gene expression and cytokine production in HL-60 promyelocytic leukaemia cells and their drug-resistant sub-clones. Biol. Trace Element Res. 2012, 149, 323–330.
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: 381
Revisions: 2 times (View History)
Update Date: 23 Apr 2023
1000/1000
ScholarVision Creations