Insulin and Insulin-like Growth Factor Signaling Pathway: History
Please note this is an old version of this entry, which may differ significantly from the current revision.
Contributor: , , , ,

Manganese (Mn) is an essential trace element, but insufficient or excessive bodily amounts can induce neurotoxicity. Mn can directly increase neuronal insulin and activate insulin-like growth factor (IGF) receptors. As an important cofactor, Mn regulates signaling pathways involved in various enzymes. The IGF signaling pathway plays a protective role in the neurotoxicity of Mn, reducing apoptosis in neurons and motor deficits by regulating its downstream protein kinase B (Akt), mitogen-activated protein kinase (MAPK), and mammalian target of rapamycin (mTOR).

  • manganese
  • neurotoxicity
  • signaling pathway
  • insulin-like growth factor (IGF)

1. Introduction

Despite concerted efforts to reduce heavy metal pollution over the past few decades, exposure to heavy metals still poses a significant threat to public health [1]. Due to natural activities or anthropogenic industrial activities, large amounts of heavy metals have been discharged into the environment. Humans are exposed to heavy metals through food, water, and air [2][3]. Heavy metals accumulate in multiple target organs, and even exposure to low levels may cause health effects [4].
Manganese (Mn) is an essential trace element involved in the synthesis and activation of many enzymes, including glutamine synthetase, pyruvate decarboxylase, manganese superoxide dismutase (MnSOD), and arginase [5]. Mn is involved in the regulation of amino acid, lipid, and carbohydrate metabolism [6]. Mn enters the human body mainly through the respiratory and digestive tracts. Under normal circumstances, an adult’s daily intake of Mn is in the order of 2–6 mg, of which 1–5% is absorbed by the duodenum. It enters the enterohepatic circulation through the portal vein and then enters the gastrointestinal tract via biliary secretions, and it is finally excreted in the feces [7].
Mn deficiency and intoxication are both associated with adverse neuropsychiatric effects, but Mn deficiency is extremely rare [8]. Excessive exposure to Mn is mainly related to occupational exposures, including welding, mining, smelters, paints, dry batteries, and Mn-rich agricultural chemicals [9][10][11]. The high concentrations of Mn in the environment are due to its abundance in the Earth’s crust, the release of Mn-containing exhaust gases and wastewater into the environment by anthropogenic activities, and the use of gasoline additives (methylcyclopentadienyl manganese tricarbonyl (MMT)) and fungicides [12][13][14][15]. Infant Mn exposure is mainly associated with excess Mn supplementation in parenteral nutrition [16].
The brain is the most sensitive target organ for Mn toxicity. Mn exists in 11 oxidation states from −3 to +7, among which Mn2+ and Mn3+ are commonly found in biological systems [17]. Mn enters the brain through the blood–brain barrier (BBB) and blood–cerebrospinal fluid barrier (BSCFB), primarily in the form of Mn2+, Mn-citrate, Mn3+-transferrin, or alpha-2- macroglobulin [18]. Mn mainly accumulates in the basal ganglia (striatum, pallidum, and substantia nigra), hippocampus, thalamus, and cortex [19]. Excessive exposure to Mn can lead to manganism, which is characterized by symptoms similar to Parkinson’s disease (PD), including gait disturbance, dystonia, and postural tremor [20]. In addition, Mn exposure has been associated with cognitive deficits, attention loss, and neuropsychological abnormalities [21]. Moreover, excessive exposure to Mn early in life can lead to learning disabilities, hyperactivity, and attention deficit disorders, and may increase the risk of neurodegenerative diseases later in life [22][23][24][25].
In addition to manganism, multiple studies demonstrated a significant association between Mn and neurodegenerative diseases. Elevated Mn levels were observed in patients with PD [26] and amyotrophic lateral sclerosis (ALS) [27], but Mn deficiency has been observed in Huntington’s disease (HD) [28], and the relationship between Mn levels and Alzheimer’s disease (AD) remains controversial [29]. However, due to the excretion of Mn, the time point and biomarkers of assessment may affect the results of these studies.
Due to the significant effects of Mn exposure on the nervous system, the mechanisms of Mn-induced neurotoxicity have been extensively studied, including neuroinflammation, protein homeostasis [30], mitochondrial function and REDOX homeostasis [31], calcium homeostasis [32], neurotransmitter metabolism [33], microRNA (miRNA) function [34], and metal homeostasis [35]. At present, the understanding of the underlying mechanisms of Mn neurotoxicity is rapidly developing. Several studies revealed the insulin-mimicking effects of Mn, which can activate several of the same metabolic kinases and directly increase peripheral and neuronal insulin and insulin-like growth factor (IGF)-1 levels in rodent models [36][37][38][39]. As an important cofactor of many kinases and phosphatases, Mn plays a key role in cell signaling pathways. Mn can activate mitogen-activated protein kinase (MAPK), protein kinase B (PKB/Akt), ataxia telangiectasia mutated (ATM), and mammalian target of rapamycin (mTOR) both in vivo and in vitro. Since these kinases regulate transcription factors, such as cyclic adenosine monophosphate (cAMP), cAMP response element-binding protein (CREB), p53, nuclear factor κB (NF-κB), and forkhead box O (FOXO), Mn can also regulate cellular function at the transcriptional level [40][41][42]. Therefore, it is important to study the role of Mn homeostasis and related signaling pathway in Mn essentiality and toxicity. However, it is not fully understood which Mn-dependent enzymes are most sensitive to changes in Mn homeostasis and the relationship between Mn and these signaling pathways.

2. Insulin and Insulin-like Growth Factor (IGF) Signaling Pathway

Insulin and IGF are homologous growth hormones that regulate cell metabolism, and their main function is to increase anabolism and reduce catabolism. Insulin has short-term metabolic effects, such as increasing glucose and amino acid transport, inducing glycolysis, glycogenesis, lipogenesis, and protein synthesis, and inhibiting gluconeogenesis, lipolysis, and protein degradation. However, IGF has chronic effects that determine cell fate, such as inducing proliferation, inhibiting apoptosis, and inducing differentiation [43]. In the brain, insulin and IGF are necessary for synaptic maintenance and activity, neurogenesis, neurite growth, and mitochondrial function [28]. Therefore, the dysregulation of these neurotrophic factors is believed to be associated with neurodegenerative diseases, and abnormal IGF/ insulin levels and insulin-related signaling changes have been observed in neurodegenerative diseases, including PD [44], AD [45], HD [28], and ALS [46].
Upon the binding of insulin or IGF receptor (IR/IGFR) on the target cell, the receptor’s tyrosine kinase is activated to phosphorylate several specific substrates, particularly insulin receptor substrates (IRSs) and Src homologous collagen (SHC) [47]. Phosphorylated tyrosine residues of these substrates are recognized by various signaling molecules containing the Src Homology 2 (SH2) domain. For example, 85 kDa regulatory subunits (p85) of phosphatidylinositol 3-kinase (PI3K), growth factor receptor binding 2 (GRB2), and protein tyrosine phosphatase 2 containing sh2 (SHP2/Syp) [48]. These bindings activate downstream signaling pathways, the PI3K pathway, and the Ras mitogen-activated protein kinase (MAPK) pathway [49]. Activation of these well-known signaling pathways is required to induce the various biological activities of IGF [50].
Mn has been shown to activate several of the same pathways as insulin/IGF, including AKT, MAPK, and mTOR, and even the IR/IGFR itself [28]. Several in vivo studies found that Mn deficiency caused glucose intolerance and reduced insulin secretion, manifested by decreased circulating IGF-1 and insulin, and increased IGFBP3 [36][37][38][39][51]. Mn supplementation improves glucose intolerance and prevents diet-induced diabetes by increasing insulin secretion and the expression of IGFR and IGF-1 in the hypothalamus, as well as increasing the expression of MnSOD [52][53][54]. These results are consistent with reports of diabetic patients responding to oral Mn therapy, as well as reduced blood Mn levels in diabetic patients [55][56].
However, few studies focused on the role of the IGF receptor itself in Mn toxicity. Tong and his colleagues found that Mn exposure reduced the expression of ATP and insulin/IGF receptors [57]. In addition, Srivastava and his colleagues found that Mn stimulated hypothalamic IGF-1 dose–dependent release and increased p-IGF-1R expression in vitro and affected hypothalamic development in adolescent rats through IGF-1/Akt/mTOR pathways [54]. Other studies demonstrated that Mn could regulate two important downstream signaling pathways of the IGF, PI3K/Akt, and MAPK signaling pathways.

2.1. PI3K/Akt Signaling Pathway

PI3K/Akt can be activated by a variety of growth factors and plays a central role in cell growth regulation, proliferation, metabolism, and cell survival, as well as neuroplasticity [58]. The interaction between the p85 and IGF-IR substrate leads to the activation of PI3K. Activated PI3K phosphorylates PIP2 to produce PIP3. PIP3, in turn, activates PDK1. PDK1 phosphorylates Akt threonine 308 residues and mTORC2 phosphorylates Akt serine 473 residues, resulting in complete Akt/PKB activation. Activated Akt phosphorylates a variety of Akt substrates, including AS160, Bad, forkhead box-containing protein, O subfamily (FoxO1), Tsc1, and glycogen synthase kinase 3β (GSK3β) [43].
PI3K inhibition reduces Mn uptake in mouse striatal cell lines (STHdh) [59]. In a Huntington’s disease cell model, exposure to both physiological (1nM) and over-physiological (10 nM) Mn levels enhanced p-IGFR/IR-dependent AKT phosphorylation, and more than 70% of Mn-induced p-Akt signaling was dependent on p-IGFR rather than other upstream and downstream effectors [60]. Researchers' previous studies have shown that exposure to toxic doses of Mn can increase the phosphorylation levels of Akt in rat hippocampal tissue [61] and PC12 cells [62]. Moreover, pretreatment of PC12 cells with LY294002, a PI3K/Akt inhibitor, further increased apoptosis, suggesting that the Mn-activated PI3K/Akt signaling pathway played a protective role. Bae et al. [63] reported that the phosphorylation level of Akt in BV2 microglia increased after 500 μM Mn treatment for 1 h. Short-term (postnatal day (PND) 8–12) exposure to Mn increased Akt phosphorylation levels in rat striatum [23]. Mn exposure also increased Akt expression levels in C. elegans [64]. However, another study reported that Mn exposure inhibited the PI3K/Akt signaling pathway in cortical neurons, and the apoptosis induced by Mn can be improved by increasing the level of the PI3K/Akt signaling pathway [65]. Brain-derived neurotrophic factor (BDNF) is a key nutrient factor involved in neurobiological mechanisms of learning and memory, and both population studies and animal experiments have confirmed that Mn exposure can reduce BDNF levels [66][67]. Mn exposure can increase α-Synuclein (α-Syn) expression in mice, and the Mn-dependent enhancement of α-Syn expression can further exacerbate the decrease in BDNF protein level and inhibit TrkB/Akt/Fyn signaling, and thus interfere with FYN-mediated phosphorylation of NMDA receptor GluN2B subtyrosine [68]. Whether Mn exposure activates or inhibits the PI3K/Akt signaling pathway may be due to different doses and/or timing of exposure and the use of different animals or cell lines, but in general, activation of the PI3K/Akt signaling pathway protects against Mn-induced neurotoxicity.
Transcription factor FOXO family proteins, substrates of Akt, can be phosphorylated by active Akt and excluded from the nucleus [69][70], thereby inhibiting the transcription of genes associated with oxidative stress protection; for example, MnSOD-2, catalase, and anti-apoptotic effects (Bim and Fas ligands) [71][72][73][74][75]. Mn exposure can increase FOXO3a levels [62][76]. In C. elegans, resistance in AKT-1/2-deficient mutant strains to Mn toxicity was higher than in wild type N2 strains, possibly because the loss of AKT-1/2 reduced the inhibition of DAF-16, and thus increased the antioxidant response [77]. In contrast, another study [40] found that treating astrocytes with 100 or 500 μM of Mn resulted in increased FOXO (dephosphorylation and phosphorylation) levels, and p-FoxO disappeared from the cytoplasm after Mn exposure, while AKT phosphorylation levels remained unchanged, but MAPK phosphorylation levels, especially p38 and ERK, and PPARγ coactivator 1 (PGC-1) levels were elevated, suggesting that FOXO may be regulated by MAPK rather than the Akt signaling pathway.

2.2. MAPK Signaling Pathway

Protein kinases ERK1/2, JNK1/2, and p38MAPK are the most important enzymes of the MAPK family [78][79]. ERK1/2 is primarily activated by growth factors and regulates gene expression, embryogenesis, proliferation, cell death/survival, and neuroplasticity [80][81]. JNK1/2/3 and p38MAPK protein kinases are commonly known as stress-activated protein kinases (SAPKs), which can be activated by cytokines and cytotoxic damage and are associated with stress and cell death [82][83]. GRB2 interacts with the IGF1 receptor substrate to activate GRB2-associated SOS guanine nucleotide exchange activity, thereby activating Ras small GTPase, which, in turn, activates MAPK. This kinase cascade is called the Ras-MAPK pathway. Activated MAPK phosphorylates transcriptional activators, leading to the induction of various IGF biological activities [43].
Acute exposure to Mn (3–6 h) at concentrations that did not affect cell viability activated MAPKs (ERK1/2 and JNK1/2) in the hippocampus and striatum sections of immature rats (PND14) [84]. Mn (500 μM) exposure increased iNOS expression in BV2 microglia, and the increase in iNOS protein expression was mediated by the JNK-ERK MAPK and PI3K/Akt signaling pathways, but not by the p38 MAPK signaling pathway [63]. Sub-acute (4 weeks) and sub-chronic (8 weeks) exposure to Mn (15 mg/kg) resulted in an increased expression of p38, p-ERK, and p-JNK in the thalamus of rats. Treatment with sodium para-aminosalicylic acid (PAS-Na), an anti-inflammatory drug, decreased p-JNK and p-P38 levels but did not decrease p-ERK levels [85]. Mn exposure also enhanced the activation of the p38 MAPK and JNK pathways in PC12 cells and mouse brain tissue, but only the activation of p38 MAPK reduced Mn-induced neuronal apoptosis through BDNF regulation [86]. Short-term exposure to Mn (PND8-12) increased the phosphorylation of DARPP-32-Thr-34, ERK1/2, and AKT in the rat striatum. TroloxTM, an antioxidant, reversed the increase in ROS and ERK1/2 phosphorylation but failed to reverse the increase in AKT phosphorylation and motor deficit induced by Mn (20 mg/kg) [23], indicating that MAPK activation is related to oxidative stress but Akt activation is not directly related to oxidative stress.
cAMP is the second intracellular messenger. CREB is a target of CAMP-dependent protein kinase A (PKA), activated by the phosphorylation of PKA at Serine-133 (Ser133), and is also regulated by Ca2+ and p38 MAPK [87][88][89][90][91]. BDNF is downstream of the cAMP-PKA-CREB signaling pathway [92]. Mn exposure increased CREB and p38 MAPK phosphorylation and apoptosis in PC12 cells, while CREB knockdown decreased BDNF levels and increased Mn-induced apoptosis, suggesting that CREB activation had a protective effect on neuronal apoptosis. In addition, the inhibition of Ca2+ and p38 MAPK significantly reduced CREB phosphorylation levels [86]. Mn exposure may also reduce the level of BDNF by inhibiting the CAMP-PKA-CREB signaling pathway in hippocampal and PC12 cells, inducing apoptosis and leading to cognitive impairment [66][93]. Phosphoprotein DARPP-32 is highly expressed in spinous neurons in the striatum. The altered phosphorylation status of Thr-34 or Thr-75 in DARPP-32 gives it the unique properties of dual function as both an inhibitor of protein phosphatase 1 (PP1) and an inhibitor of PKA. Rats exposed to 5 or 10 mg Mn/kg on PND8-12 showed increased phosphorylation of DARPP-32 at Thr-34 in the striatum on PND14 [23].

This entry is adapted from the peer-reviewed paper 10.3390/cells12242842

References

  1. Grandjean, P.; Barouki, R.; Bellinger, D.C.; Casteleyn, L.; Chadwick, L.H.; Cordier, S.; Etzel, R.A.; Gray, K.A.; Ha, E.H.; Junien, C.; et al. Life-Long Implications of Developmental Exposure to Environmental Stressors: New Perspectives. Endocrinology 2015, 156, 3408–3415.
  2. Al Osman, M.; Yang, F.; Massey, I.Y. Exposure routes and health effects of heavy metals on children. Biometals 2019, 32, 563–573.
  3. Tan, S.Y.; Praveena, S.M.; Abidin, E.Z.; Cheema, M.S. A review of heavy metals in indoor dust and its human health-risk implications. Rev. Environ. Health 2016, 31, 447–456.
  4. Fu, Z.; Xi, S. The effects of heavy metals on human metabolism. Toxicol. Mech. Methods 2020, 30, 167–176.
  5. Horning, K.J.; Caito, S.W.; Tipps, K.G.; Bowman, A.B.; Aschner, M. Manganese Is Essential for Neuronal Health. Annu. Rev. Nutr. 2015, 35, 71–108.
  6. Wedler, F.C. Biological significance of manganese in mammalian systems. Prog. Med. Chem. 1993, 30, 89–133.
  7. Davis, C.D.; Zech, L.; Greger, J.L. Manganese metabolism in rats: An improved methodology for assessing gut endogenous losses. Proc. Soc. Exp. Biol. Med. 1993, 202, 103–108.
  8. Vollet, K.; Haynes, E.N.; Dietrich, K.N. Manganese Exposure and Cognition Across the Lifespan: Contemporary Review and Argument for Biphasic Dose-Response Health Effects. Curr. Environ. Health Rep. 2016, 3, 392–404.
  9. Blanc, P.D. The early history of manganese and the recognition of its neurotoxicity, 1837-1936. Neurotoxicology 2018, 64, 5–11.
  10. Boudissa, S.M.; Lambert, J.; Muller, C.; Kennedy, G.; Gareau, L.; Zayed, J. Manganese concentrations in the soil and air in the vicinity of a closed manganese alloy production plant. Sci. Total Environ. 2006, 361, 67–72.
  11. Jiang, F.; Ren, B.; Hursthouse, A.S.; Zhou, Y. Trace Metal Pollution in Topsoil Surrounding the Xiangtan Manganese Mine Area (South-Central China): Source Identification, Spatial Distribution and Assessment of Potential Ecological Risks. Int. J. Environ. Res. Public Health 2018, 15, 2412.
  12. Gulson, B.; Mizon, K.; Taylor, A.; Korsch, M.; Stauber, J.; Davis, J.M.; Louie, H.; Wu, M.; Swan, H. Changes in manganese and lead in the environment and young children associated with the introduction of methylcyclopentadienyl manganese tricarbonyl in gasoline--preliminary results. Environ. Res. 2006, 100, 100–114.
  13. Herndon, E.M.; Jin, L.; Brantley, S.L. Soils reveal widespread manganese enrichment from industrial inputs. Environ. Sci. Technol. 2011, 45, 241–247.
  14. Kullar, S.S.; Shao, K.; Surette, C.; Foucher, D.; Mergler, D.; Cormier, P.; Bellinger, D.C.; Barbeau, B.; Sauve, S.; Bouchard, M.F. A benchmark concentration analysis for manganese in drinking water and IQ deficits in children. Environ. Int. 2019, 130, 104889.
  15. Solis-Vivanco, R.; Rodriguez-Agudelo, Y.; Riojas-Rodriguez, H.; Rios, C.; Rosas, I.; Montes, S. Cognitive impairment in an adult Mexican population non-occupationally exposed to manganese. Environ. Toxicol. Pharmacol. 2009, 28, 172–178.
  16. Reynolds, A.P.; Kiely, E.; Meadows, N. Manganese in long term paediatric parenteral nutrition. Arch. Dis. Child. 1994, 71, 527–528.
  17. Stephenson, A.P.; Schneider, J.A.; Nelson, B.C.; Atha, D.H.; Jain, A.; Soliman, K.F.; Aschner, M.; Mazzio, E.; Renee Reams, R. Manganese-induced oxidative DNA damage in neuronal SH-SY5Y cells: Attenuation of thymine base lesions by glutathione and N-acetylcysteine. Toxicol. Lett. 2013, 218, 299–307.
  18. Baj, J.; Flieger, W.; Barbachowska, A.; Kowalska, B.; Flieger, M.; Forma, A.; Teresinski, G.; Portincasa, P.; Buszewicz, G.; Radzikowska-Buchner, E.; et al. Consequences of Disturbing Manganese Homeostasis. Int. J. Mol. Sci. 2023, 24, 14959.
  19. Tuschl, K.; Mills, P.B.; Clayton, P.T. Manganese and the brain. Int. Rev. Neurobiol. 2013, 110, 277–312.
  20. Perl, D.P.; Olanow, C.W. The neuropathology of manganese-induced Parkinsonism. J. Neuropathol. Exp. Neurol. 2007, 66, 675–682.
  21. Guilarte, T.R.; Burton, N.C.; Verina, T.; Prabhu, V.V.; Becker, K.G.; Syversen, T.; Schneider, J.S. Increased APLP1 expression and neurodegeneration in the frontal cortex of manganese-exposed non-human primates. J. Neurochem. 2008, 105, 1948–1959.
  22. Bjorklund, G.; Chartrand, M.S.; Aaseth, J. Manganese exposure and neurotoxic effects in children. Environ. Res. 2017, 155, 380–384.
  23. Cordova, F.M.; Aguiar, A.S., Jr.; Peres, T.V.; Lopes, M.W.; Goncalves, F.M.; Remor, A.P.; Lopes, S.C.; Pilati, C.; Latini, A.S.; Prediger, R.D.; et al. In vivo manganese exposure modulates Erk, Akt and Darpp-32 in the striatum of developing rats, and impairs their motor function. PLoS ONE 2012, 7, e33057.
  24. Rodrigues, J.L.G.; Araujo, C.F.S.; Dos Santos, N.R.; Bandeira, M.J.; Anjos, A.L.S.; Carvalho, C.F.; Lima, C.S.; Abreu, J.N.S.; Mergler, D.; Menezes-Filho, J.A. Airborne manganese exposure and neurobehavior in school-aged children living near a ferro-manganese alloy plant. Environ. Res. 2018, 167, 66–77.
  25. Shih, J.H.; Zeng, B.Y.; Lin, P.Y.; Chen, T.Y.; Chen, Y.W.; Wu, C.K.; Tseng, P.T.; Wu, M.K. Association between peripheral manganese levels and attention-deficit/hyperactivity disorder: A preliminary meta-analysis. Neuropsychiatr. Dis. Treat. 2018, 14, 1831–1842.
  26. Du, K.; Liu, M.Y.; Pan, Y.Z.; Zhong, X.; Wei, M.J. Association of circulating manganese levels with Parkinson’s disease: A meta-analysis. Neurosci. Lett. 2018, 665, 92–98.
  27. Roos, P.M.; Lierhagen, S.; Flaten, T.P.; Syversen, T.; Vesterberg, O.; Nordberg, M. Manganese in cerebrospinal fluid and blood plasma of patients with amyotrophic lateral sclerosis. Exp. Biol. Med. 2012, 237, 803–810.
  28. Bryan, M.R.; Bowman, A.B. Manganese and the Insulin-IGF Signaling Network in Huntington’s Disease and Other Neurodegenerative Disorders. Adv. Neurobiol. 2017, 18, 113–142.
  29. Du, K.; Liu, M.; Pan, Y.; Zhong, X.; Wei, M. Association of Serum Manganese Levels with Alzheimer’s Disease and Mild Cognitive Impairment: A Systematic Review and Meta-Analysis. Nutrients 2017, 9, 231.
  30. Harischandra, D.S.; Ghaisas, S.; Zenitsky, G.; Jin, H.; Kanthasamy, A.; Anantharam, V.; Kanthasamy, A.G. Manganese-Induced Neurotoxicity: New Insights Into the Triad of Protein Misfolding, Mitochondrial Impairment, and Neuroinflammation. Front. Neurosci. 2019, 13, 654.
  31. Martinez-Finley, E.J.; Gavin, C.E.; Aschner, M.; Gunter, T.E. Manganese neurotoxicity and the role of reactive oxygen species. Free Radic. Biol. Med. 2013, 62, 65–75.
  32. Ijomone, O.M.; Aluko, O.M.; Okoh, C.O.A.; Martins, A.C., Jr.; Aschner, M. Role for calcium signaling in manganese neurotoxicity. J. Trace Elem. Med. Biol. 2019, 56, 146–155.
  33. Soares, A.T.G.; Silva, A.C.; Tinkov, A.A.; Khan, H.; Santamaria, A.; Skalnaya, M.G.; Skalny, A.V.; Tsatsakis, A.; Bowman, A.B.; Aschner, M.; et al. The impact of manganese on neurotransmitter systems. J. Trace Elem. Med. Biol. 2020, 61, 126554.
  34. Wallace, D.R.; Taalab, Y.M.; Heinze, S.; Tariba Lovakovic, B.; Pizent, A.; Renieri, E.; Tsatsakis, A.; Farooqi, A.A.; Javorac, D.; Andjelkovic, M.; et al. Toxic-Metal-Induced Alteration in miRNA Expression Profile as a Proposed Mechanism for Disease Development. Cells 2020, 9, 901.
  35. Chen, P.; Bornhorst, J.; Aschner, M. Manganese metabolism in humans. Front. Biosci. 2018, 23, 1655–1679.
  36. Baly, D.L.; Curry, D.L.; Keen, C.L.; Hurley, L.S. Effect of manganese deficiency on insulin secretion and carbohydrate homeostasis in rats. J. Nutr. 1984, 114, 1438–1446.
  37. Baly, D.L.; Lee, I.; Doshi, R. Mechanism of decreased insulinogenesis in manganese-deficient rats. Decreased insulin mRNA levels. FEBS Lett. 1988, 239, 55–58.
  38. Baly, D.L.; Schneiderman, J.S.; Garcia-Welsh, A.L. Effect of manganese deficiency on insulin binding, glucose transport and metabolism in rat adipocytes. J. Nutr. 1990, 120, 1075–1079.
  39. Hiney, J.K.; Srivastava, V.K.; Dees, W.L. Manganese induces IGF-1 and cyclooxygenase-2 gene expressions in the basal hypothalamus during prepubertal female development. Toxicol. Sci. 2011, 121, 389–396.
  40. Exil, V.; Ping, L.; Yu, Y.; Chakraborty, S.; Caito, S.W.; Wells, K.S.; Karki, P.; Lee, E.; Aschner, M. Activation of MAPK and FoxO by manganese (Mn) in rat neonatal primary astrocyte cultures. PLoS ONE 2014, 9, e94753.
  41. Jang, B.C. Induction of COX-2 in human airway cells by manganese: Role of PI3K/PKB, p38 MAPK, PKCs, Src, and glutathione depletion. Toxicol. Vitr. 2009, 23, 120–126.
  42. Moreno, J.A.; Streifel, K.M.; Sullivan, K.A.; Hanneman, W.H.; Tjalkens, R.B. Manganese-induced NF-kappaB activation and nitrosative stress is decreased by estrogen in juvenile mice. Toxicol. Sci. 2011, 122, 121–133.
  43. Hakuno, F.; Takahashi, S.I. IGF1 receptor signaling pathways. J. Mol. Endocrinol. 2018, 61, T69–T86.
  44. Ma, J.; Jiang, Q.; Xu, J.; Sun, Q.; Qiao, Y.; Chen, W.; Wu, Y.; Wang, Y.; Xiao, Q.; Liu, J.; et al. Plasma insulin-like growth factor 1 is associated with cognitive impairment in Parkinson’s disease. Dement. Geriatr. Cogn. Disord. 2015, 39, 251–256.
  45. De la Monte, S.M.; Wands, J.R. Alzheimer’s disease is type 3 diabetes-evidence reviewed. J. Diabetes Sci. Technol. 2008, 2, 1101–1113.
  46. Bilic, E.; Bilic, E.; Rudan, I.; Kusec, V.; Zurak, N.; Delimar, D.; Zagar, M. Comparison of the growth hormone, IGF-1 and insulin in cerebrospinal fluid and serum between patients with motor neuron disease and healthy controls. Eur. J. Neurol. 2006, 13, 1340–1345.
  47. Craparo, A.; O’Neill, T.J.; Gustafson, T.A. Non-SH2 domains within insulin receptor substrate-1 and SHC mediate their phosphotyrosine-dependent interaction with the NPEY motif of the insulin-like growth factor I receptor. J. Biol. Chem. 1995, 270, 15639–15643.
  48. Copps, K.D.; White, M.F. Regulation of insulin sensitivity by serine/threonine phosphorylation of insulin receptor substrate proteins IRS1 and IRS2. Diabetologia 2012, 55, 2565–2582.
  49. Jones, J.I.; Clemmons, D.R. Insulin-like growth factors and their binding proteins: Biological actions. Endocr. Rev. 1995, 16, 3–34.
  50. Hakuno, F.; Fukushima, T.; Yoneyama, Y.; Kamei, H.; Ozoe, A.; Yoshihara, H.; Yamanaka, D.; Shibano, T.; Sone-Yonezawa, M.; Yu, B.C.; et al. The Novel Functions of High-Molecular-Mass Complexes Containing Insulin Receptor Substrates in Mediation and Modulation of Insulin-Like Activities: Emerging Concept of Diverse Functions by IRS-Associated Proteins. Front. Endocrinol. 2015, 6, 73.
  51. Clegg, M.S.; Donovan, S.M.; Monaco, M.H.; Baly, D.L.; Ensunsa, J.L.; Keen, C.L. The influence of manganese deficiency on serum IGF-1 and IGF binding proteins in the male rat. Proc. Soc. Exp. Biol. Med. 1998, 219, 41–47.
  52. Dearth, R.K.; Hiney, J.K.; Srivastava, V.K.; Hamilton, A.M.; Dees, W.L. Prepubertal exposure to elevated manganese results in estradiol regulated mammary gland ductal differentiation and hyperplasia in female rats. Exp. Biol. Med. 2014, 239, 871–882.
  53. Lee, S.H.; Jouihan, H.A.; Cooksey, R.C.; Jones, D.; Kim, H.J.; Winge, D.R.; McClain, D.A. Manganese supplementation protects against diet-induced diabetes in wild type mice by enhancing insulin secretion. Endocrinology 2013, 154, 1029–1038.
  54. Srivastava, V.K.; Hiney, J.K.; Dees, W.L. Manganese-Stimulated Kisspeptin Is Mediated by the IGF-1/Akt/Mammalian Target of Rapamycin Pathway in the Prepubertal Female Rat. Endocrinology 2016, 157, 3233–3241.
  55. Ekmekcioglu, C.; Prohaska, C.; Pomazal, K.; Steffan, I.; Schernthaner, G.; Marktl, W. Concentrations of seven trace elements in different hematological matrices in patients with type 2 diabetes as compared to healthy controls. Biol. Trace Elem. Res. 2001, 79, 205–219.
  56. Koh, E.S.; Kim, S.J.; Yoon, H.E.; Chung, J.H.; Chung, S.; Park, C.W.; Chang, Y.S.; Shin, S.J. Association of blood manganese level with diabetes and renal dysfunction: A cross-sectional study of the Korean general population. BMC Endocr. Disord. 2014, 14, 24.
  57. Tong, M.; Dong, M.; de la Monte, S.M. Brain insulin-like growth factor and neurotrophin resistance in Parkinson’s disease and dementia with Lewy bodies: Potential role of manganese neurotoxicity. J. Alzheimers Dis. 2009, 16, 585–599.
  58. Van der Heide, L.P.; Ramakers, G.M.; Smidt, M.P. Insulin signaling in the central nervous system: Learning to survive. Prog. Neurobiol. 2006, 79, 205–221.
  59. Bryan, M.R.; Uhouse, M.A.; Nordham, K.D.; Joshi, P.; Rose, D.I.R.; O’Brien, M.T.; Aschner, M.; Bowman, A.B. Phosphatidylinositol 3 kinase (PI3K) modulates manganese homeostasis and manganese-induced cell signaling in a murine striatal cell line. Neurotoxicology 2018, 64, 185–194.
  60. Bryan, M.R.; Nordham, K.D.; Rose, D.I.R.; O’Brien, M.T.; Joshi, P.; Foshage, A.M.; Goncalves, F.M.; Nitin, R.; Uhouse, M.A.; Aschner, M.; et al. Manganese Acts upon Insulin/IGF Receptors to Phosphorylate AKT and Increase Glucose Uptake in Huntington’s Disease Cells. Mol. Neurobiol. 2020, 57, 1570–1593.
  61. Cheng, H.; Xia, B.; Su, C.; Chen, K.; Chen, X.; Chen, P.; Zou, Y.; Yang, X. PI3K/Akt signaling pathway and Hsp70 activate in hippocampus of rats with chronic manganese sulfate exposure. J. Trace Elem. Med. Biol. 2018, 50, 332–338.
  62. Tan, Y.; Cheng, H.; Su, C.; Chen, P.; Yang, X. PI3K/Akt Signaling Pathway Ameliorates Oxidative Stress-Induced Apoptosis upon Manganese Exposure in PC12 Cells. Biol. Trace Elem. Res. 2022, 200, 749–760.
  63. Bae, J.H.; Jang, B.C.; Suh, S.I.; Ha, E.; Baik, H.H.; Kim, S.S.; Lee, M.Y.; Shin, D.H. Manganese induces inducible nitric oxide synthase (iNOS) expression via activation of both MAP kinase and PI3K/Akt pathways in BV2 microglial cells. Neurosci. Lett. 2006, 398, 151–154.
  64. Avila, D.S.; Somlyai, G.; Somlyai, I.; Aschner, M. Anti-aging effects of deuterium depletion on Mn-induced toxicity in a C. elegans model. Toxicol. Lett. 2012, 211, 319–324.
  65. Yang, Y.; Wei, F.; Wang, J.; Chen, R.; Zhang, J.; Li, D.; Gan, D.; Yang, X.; Zou, Y. Manganese modifies Neurotrophin-3 (NT3) and its tropomyosin receptor kinase C (TrkC) in the cortex: Implications for manganese-induced neurotoxicity. Food Chem. Toxicol. 2020, 135, 110925.
  66. Liang, G.; Qin, H.; Zhang, L.; Ma, S.; Huang, X.; Lv, Y.; Qing, L.; Li, Q.; Xiong, Y.; Huang, Y.; et al. Effects of chronic manganese exposure on the learning and memory of rats by observing the changes in the hippocampal cAMP signaling pathway. Food Chem. Toxicol. 2015, 83, 261–267.
  67. Zou, Y.; Qing, L.; Zeng, X.; Shen, Y.; Zhong, Y.; Liu, J.; Li, Q.; Chen, K.; Lv, Y.; Huang, D.; et al. Cognitive function and plasma BDNF levels among manganese-exposed smelters. Occup. Environ. Med. 2014, 71, 189–194.
  68. Ma, Z.; Liu, K.; Li, X.R.; Wang, C.; Liu, C.; Yan, D.Y.; Deng, Y.; Liu, W.; Xu, B. Alpha-synuclein is involved in manganese-induced spatial memory and synaptic plasticity impairments via TrkB/Akt/Fyn-mediated phosphorylation of NMDA receptors. Cell Death Dis. 2020, 11, 834.
  69. Alvarez, B.; Martinez, A.C.; Burgering, B.M.; Carrera, A.C. Forkhead transcription factors contribute to execution of the mitotic programme in mammals. Nature 2001, 413, 744–747.
  70. Brunet, A.; Bonni, A.; Zigmond, M.J.; Lin, M.Z.; Juo, P.; Hu, L.S.; Anderson, M.J.; Arden, K.C.; Blenis, J.; Greenberg, M.E. Akt promotes cell survival by phosphorylating and inhibiting a Forkhead transcription factor. Cell 1999, 96, 857–868.
  71. Al-Mubarak, B.; Soriano, F.X.; Hardingham, G.E. Synaptic NMDAR activity suppresses FOXO1 expression via a cis-acting FOXO binding site: FOXO1 is a FOXO target gene. Channels 2009, 3, 233–238.
  72. Birkenkamp, K.U.; Coffer, P.J. FOXO transcription factors as regulators of immune homeostasis: Molecules to die for? J. Immunol. 2003, 171, 1623–1629.
  73. Kops, G.J.; Dansen, T.B.; Polderman, P.E.; Saarloos, I.; Wirtz, K.W.; Coffer, P.J.; Huang, T.T.; Bos, J.L.; Medema, R.H.; Burgering, B.M. Forkhead transcription factor FOXO3a protects quiescent cells from oxidative stress. Nature 2002, 419, 316–321.
  74. Nemoto, S.; Finkel, T. Redox regulation of forkhead proteins through a p66shc-dependent signaling pathway. Science 2002, 295, 2450–2452.
  75. Van Der Heide, L.P.; Hoekman, M.F.; Smidt, M.P. The ins and outs of FoxO shuttling: Mechanisms of FoxO translocation and transcriptional regulation. Biochem. J. 2004, 380, 297–309.
  76. Zhao, X.; Liu, Y.; Zhu, G.; Liang, Y.; Liu, B.; Wu, Y.; Han, M.; Sun, W.; Han, Y.; Chen, G.; et al. SIRT1 downregulation mediated Manganese-induced neuronal apoptosis through activation of FOXO3a-Bim/PUMA axis. Sci. Total Environ. 2019, 646, 1047–1055.
  77. Peres, T.V.; Arantes, L.P.; Miah, M.R.; Bornhorst, J.; Schwerdtle, T.; Bowman, A.B.; Leal, R.B.; Aschner, M. Role of Caenorhabditis elegans AKT-1/2 and SGK-1 in Manganese Toxicity. Neurotox. Res. 2018, 34, 584–596.
  78. Chang, L.; Karin, M. Mammalian MAP kinase signalling cascades. Nature 2001, 410, 37–40.
  79. Morrison, D.K. MAP kinase pathways. Cold Spring Harb. Perspect. Biol. 2012, 4, a011254.
  80. Chen, Z.; Gibson, T.B.; Robinson, F.; Silvestro, L.; Pearson, G.; Xu, B.; Wright, A.; Vanderbilt, C.; Cobb, M.H. MAP kinases. Chem. Rev. 2001, 101, 2449–2476.
  81. Thomas, G.M.; Huganir, R.L. MAPK cascade signalling and synaptic plasticity. Nat. Rev. Neurosci. 2004, 5, 173–183.
  82. Cowan, K.J.; Storey, K.B. Mitogen-activated protein kinases: New signaling pathways functioning in cellular responses to environmental stress. J. Exp. Biol. 2003, 206, 1107–1115.
  83. Mielke, K.; Herdegen, T. JNK and p38 stresskinases--degenerative effectors of signal-transduction-cascades in the nervous system. Prog. Neurobiol. 2000, 61, 45–60.
  84. Peres, T.V.; Pedro, D.Z.; de Cordova, F.M.; Lopes, M.W.; Goncalves, F.M.; Mendes-de-Aguiar, C.B.; Walz, R.; Farina, M.; Aschner, M.; Leal, R.B. In vitro manganese exposure disrupts MAPK signaling pathways in striatal and hippocampal slices from immature rats. Biomed. Res. Int. 2013, 2013, 769295.
  85. Deng, Y.; Peng, D.; Yang, C.; Zhao, L.; Li, J.; Lu, L.; Zhu, X.; Li, S.; Aschner, M.; Jiang, Y. Preventive treatment with sodium para-aminosalicylic acid inhibits manganese-induced apoptosis and inflammation via the MAPK pathway in rat thalamus. Drug Chem. Toxicol. 2023, 46, 59–68.
  86. Zhu, G.; Liu, Y.; Zhi, Y.; Jin, Y.; Li, J.; Shi, W.; Liu, Y.; Han, Y.; Yu, S.; Jiang, J.; et al. PKA- and Ca2+-dependent p38 MAPK/CREB activation protects against manganese-mediated neuronal apoptosis. Toxicol. Lett. 2019, 309, 10–19.
  87. Benito, E.; Valor, L.M.; Jimenez-Minchan, M.; Huber, W.; Barco, A. cAMP response element-binding protein is a primary hub of activity-driven neuronal gene expression. J. Neurosci. 2011, 31, 18237–18250.
  88. Impey, S.; Obrietan, K.; Storm, D.R. Making new connections: Role of ERK/MAP kinase signaling in neuronal plasticity. Neuron 1999, 23, 11–14.
  89. Mayr, B.; Montminy, M. Transcriptional regulation by the phosphorylation-dependent factor CREB. Nat. Rev. Mol. Cell Biol. 2001, 2, 599–609.
  90. Xing, J.; Kornhauser, J.M.; Xia, Z.; Thiele, E.A.; Greenberg, M.E. Nerve growth factor activates extracellular signal-regulated kinase and p38 mitogen-activated protein kinase pathways to stimulate CREB serine 133 phosphorylation. Mol. Cell Biol. 1998, 18, 1946–1955.
  91. Yu, S.; Cheng, Q.; Li, L.; Liu, M.; Yang, Y.; Ding, F. 2-(4-Methoxyphenyl)ethyl-2-acetamido-2-deoxy-beta-d-pyranoside confers neuroprotection in cell and animal models of ischemic stroke through calpain1/PKA/CREB-mediated induction of neuronal glucose transporter 3. Toxicol. Appl. Pharmacol. 2014, 277, 259–269.
  92. Tao, X.; Finkbeiner, S.; Arnold, D.B.; Shaywitz, A.J.; Greenberg, M.E. Ca2+ influx regulates BDNF transcription by a CREB family transcription factor-dependent mechanism. Neuron 1998, 20, 709–726.
  93. Yang, Y.; Ma, S.; Wei, F.; Liang, G.; Yang, X.; Huang, Y.; Wang, J.; Zou, Y. Pivotal role of cAMP-PKA-CREB signaling pathway in manganese-induced neurotoxicity in PC12 cells. Environ. Toxicol. 2019, 34, 1052–1062.
More
This entry is offline, you can click here to edit this entry!
Video Production Service