EFFECTS OF MOUNJARO ON BRAIN CELLS

REGISTRO DOI: 10.69849/revistaft/dt10202601151251


Diego Santiago Marinho¹
Antônio Levy Carvalho Nobre¹
Vicente Bruno de Freitas Guimarães¹
Carlos Otávio de Arruda Bezerra Filho¹
Bruna Mara Machado Ribeiro2


Abstract 

Mounjaro works by activating two gut hormone receptors, GIP and GLP-1, to manage  blood sugar. This stimulates insulin release when glucose is high, reduces liver glucagon  production, slows stomach emptying to control post-meal sugar spikes, and increases  fullness to reduce appetite and food intake. Mounjaro with reported effects on central  nervous system neurotransmission, synaptic plasticity, and neuroprotection. Here, we  investigated the potential anti-inflammatory and neuroprotective effects of Mounjaro in  primary hippocampal neuronal cultures exposed to the viral mimetic  polyinosinic:polycytidylic acid (poly(I:C)). Primary hippocampal neurons were treated  with Mounjaro (1.5 µM or 15 µM) or risperidone (12 µM or 23 µM) in the presence or  absence of poly(I:C) (100 µg/mL) for 48 h. Cell viability and cytotoxicity were assessed  by MTT and LDH assays, respectively. Inflammatory and neuronal markers were  evaluated by immunofluorescence (iNOS, NFκB p50, DCX, and NeuN) and Western  blotting (NFκB p65). Culture supernatants were analyzed for IL-6, nitrite, and BDNF.  Poly(I:C) reduced cell viability and increased iNOS expression, NFκB activation  (p50/p65), IL-6, and nitrite, while decreasing BDNF levels and DCX immunoexpression.  Treatment with Mounjaro (15 µM) or risperidone (23 µM) significantly attenuated the  poly(I:C)-induced increases in iNOS, NFκB (p50/p65), IL-6, and nitrite. Mounjaro also  prevented the poly(I:C)-induced reduction in BDNF. Together, these findings indicate  that Mounjaro and risperidone exert anti-inflammatory and neuroprotective effects in  primary hippocampal neurons under poly(I:C)-induced inflammatory challenge. 

Keywords: Mounjaro; poly(I:C); hippocampus; primary culture, Brain. 

1. Introduction 

Mounjaro is an incretin-based drug that has been associated with central effects on  neurotransmission and, potentially, synaptic plasticity and neuroprotection. Based on  these reports, the present study was designed to further investigate the neuroprotective  potential of Mounjaro in primary hippocampal neuronal cultures subjected to an  inflammatory stimulus induced by exposure to polyinosinic:polycytidylic acid (poly(I:C)). Poly(I:C) is a synthetic analog of double-stranded RNA widely used to  mimic the innate immune response to viral infection and to study neuroimmune  mechanisms implicated in neuropsychiatric and neurodevelopmental disorders. 

In mammalian systems, poly(I:C) is recognized by toll-like receptor 3 (TLR3), leading to  the induction of pro-inflammatory mediators, including interleukin (IL)-1β, IL-6, and  tumor necrosis factor (TNF)-α (Alexopoulou et al., 2001). Poly(I:C) is also a strong  inducer of type I interferons, such as IFNα and IFNβ (Lafon et al., 2006). Consequently,  poly(I:C) immune challenge has gained attention as a model to study the development of  a “disease-like brain” after early-life immune activation (Meyer, 2013). 

Experimental evidence suggests that pharmacological interventions with antipsychotics  or antidepressants during adolescence may block the emergence of behavioral alterations  induced by prenatal poly(I:C) exposure (Meyer et al., 2010). For example, early  administration of risperidone prevented brain morphological changes such as lateral  ventricle enlargement and hippocampal atrophy in a poly(I:C) model (Piontkewitz et al.,  2009). However, antipsychotics can cause significant adverse effects (e.g.,  extrapyramidal symptoms, sedation, hyperprolactinemia, and weight gain) and have been  associated with an increased risk of diabetes in children and adolescents (Kumra et al.,  2007). Therefore, safer preventive strategies are needed for translational application  (Sommer et al., 2016). 

Several studies have highlighted an important role for intestinal incretins, particularly  glucagon-like peptide-1 (GLP-1), in brain function and neuronal survival. GLP-1  receptors contribute to synaptic plasticity and cognitive processes; in GLP-1 receptor  knockout mice, hippocampal synaptic plasticity is markedly impaired (Abbas et al.,  2009). GLP-1 analogues can modulate neurotransmitter release and promote synaptic  plasticity, supporting their neuroprotective properties in preclinical models (Gault and  Hölscher, 2008). 

The dentate gyrus (DG) of the hippocampus is a key neurogenic niche in the adult brain.  The molecular pathways controlling adult hippocampal neurogenesis remain  incompletely understood. Incretin-related mechanisms may represent potential regulators  of hippocampal neurogenesis and neuroprotection (Nyberg et al., 2005). In this context,  we evaluated whether Mounjaro could mitigate poly(I:C)-induced neuroinflammatory  changes and preserve neuronal markers in primary hippocampal neuronal cultures.

2. Materials and Methods 

2.1 Animals 

Newborn mice (postnatal day 1) from different litters were used. All procedures complied  with the Brazilian College of Animal Experimentation (COBEA) guidelines and the NIH  Guide for the Care and Use of Laboratory Animals. The Ethics and Animal Research  Committee of the Federal University of Ceará approved the study. 

2.2 Drugs and reagents 

Primary hippocampal neuronal cultures were exposed to Mounjaro (1.5 µM or 15 µM) or  risperidone (12 µM or 23 µM) in the presence or absence of poly(I:C) (100 µg/mL) for  48 h. Poly(I:C) and other chemicals were purchased from Sigma-Aldrich (St. Louis, MO,  USA). Culture media components were obtained from Thermo Fisher Scientific  (Waltham, MA, USA). 

2.3 Primary hippocampal neuronal cultures 

On postnatal day 1, pups were anesthetized with pentobarbital (50 mg/kg, i.p.) and  euthanized by decapitation. The hippocampi from both hemispheres were dissected under  a Leica binocular microscope. Tissue was gently dissociated mechanically with a sterile  pipette in Neurobasal medium containing 0.03% trypsin and incubated for 20 min at 37  °C in 5% CO₂. Cells were then suspended in Neurobasal medium supplemented with 1%  fetal bovine serum (FBS), 1% human serum (HS), 2% B27 supplement, 0.25% L glutamine, and 50 U/mL penicillin–streptomycin (control condition). Cells were plated at  a density of 1.5 × 10⁵ cells/cm² onto poly-D-lysine–coated (50 µg/mL) 12-mm round  coverslips in 96-well plates (for the MTT assay). Cultures were maintained at 37 °C in a  humidified atmosphere containing 5% CO₂. Under these conditions, glial cell growth was  reduced to <0.5%, yielding a nearly pure neuronal population (Brewer et al., 1993;  Kyoung et al., 2014). 

2.4 Drug treatment and culture-medium collection 

Primary hippocampal neuronal cultures were treated with Mounjaro (1.5 µM or 15 µM)  or risperidone (12 µM or 23 µM) in the presence or absence of poly(I:C) (100 µg/mL) for  48 h and then subjected to viability and biochemical assays. Control cells were cultured  under the same conditions without test drugs. For culture-medium collection,  supernatants were harvested and centrifuged at 1000 × g for 5 min to remove debris. 

2.5 MTT assay 

Cell viability was assessed by measuring the reduction of MTT by mitochondrial  dehydrogenases, as previously described (Pusceddu et al., 2015). Culture medium was  replaced with 300 µL of fresh medium containing MTT (1 mg/mL) and incubated for 3 h  at 37 °C. Formazan crystals were dissolved in 100 µL dimethyl sulfoxide (DMSO), and absorbance was read at 570 nm using a microplate reader. Results are expressed as  percentage viability relative to untreated controls (100%). 

2.6 LDH assay 

LDH release was used as an index of cytotoxicity. LDH activity was measured using an  enzymatic coupled reaction (lactate/NAD⁺/diaphorase and a redox dye), following the  manufacturer’s instructions. Fluorescence was measured with excitation at 560 nm and  emission at 590 nm after a short incubation (10–20 min). 

2.7 Immunofluorescence 

Cells were fixed with 2.5% paraformaldehyde in phosphate-buffered saline (PBS) for 25  min at 4 °C, washed, and permeabilized in PBS containing 0.02% Tween 20 (three  washes, 5 min each). Non-specific binding was blocked with 5% normal bovine serum in  PBS/Tween overnight at 4 °C. Cells were incubated overnight at 4 °C with primary  antibodies against inducible nitric oxide synthase (iNOS; 1:200, goat polyclonal), NFκB  p50 (1:200, rabbit polyclonal), doublecortin (DCX; 1:200, rabbit polyclonal), or Alexa  Fluor 488–conjugated anti-NeuN (1:200, mouse monoclonal) (all from Sigma-Aldrich).  After washing, cells were incubated with appropriate secondary antibodies (Alexa Fluor  568 donkey anti-rabbit IgG or Alexa Fluor 488 donkey anti-goat IgG; 1:200). Nuclei  were counterstained with DAPI. Images were acquired on an Olympus FV1000 confocal  microscope (20× and 60×). Quantification of immunoreactive area was performed using  ImageJ. 

2.8 Western blotting 

Cells were washed with ice-cold PBS and lysed in RIPA buffer (25 mM Tris-HCl, pH  7.6; 150 mM NaCl; 5 mM EDTA; 1% Triton X-100; 1% sodium deoxycholate; 0.1%  SDS) containing protease inhibitors. Lysates were sonicated twice for 5 s and cleared by  centrifugation (12,000 rpm, 15 min, 4 °C). Protein concentration was determined by the  BCA method. Samples (20 µg protein) were separated by 10% SDS-PAGE and  transferred to PVDF membranes. Membranes were blocked with 5% BSA for 1 h and  incubated overnight with rabbit anti-NFκB p65 (1:200) or mouse anti-α-tubulin (1:4000)  primary antibodies. After washing, membranes were incubated with HRP-conjugated  secondary antibodies (1:1000) for 90 min at room temperature. Bands were visualized  using ECL and quantified by densitometry in ImageJ (n = 4/group). 

2.9 IL-6 and BDNF measurements 

IL-6 and BDNF levels in culture supernatants were measured by enzyme immunoassay  according to the manufacturer’s instructions (R&D Systems, Minneapolis, MN, USA).  Results are expressed as pg/mL of culture medium.

2.10 Nitrite determination 

Nitrite levels, an indirect measure of nitric oxide (NO) production, were determined in  culture supernatants using the Griess reaction (Green et al., 1981). Results are expressed  as nM/mL of culture medium. 

2.11 Statistical analysis 

Data were analyzed using the Kruskal–Wallis one-way analysis of variance followed by  Dunn’s multiple-comparisons test. Normality was assessed with the D’Agostino–Pearson  omnibus test. Results are expressed as mean ± standard error of the mean (SEM). P  values ≤ 0.05 were considered statistically significant. Analyses were performed in  GraphPad Prism (version 6.0, Windows). 

3. Results 

3.1 Mounjaro and risperidone protect hippocampal neurons from poly(I:C)- induced loss of viability 

The MTT assay showed that poly(I:C) significantly reduced neuronal viability compared with control cultures (P < 0.05). This reduction was prevented by Mounjaro (15 µM) or  risperidone (23 µM) (Figure 1).

Figure 1. Primary hippocampal neuronal cultures were exposed to Mounjaro (1.5 µM or  15 µM) or risperidone (12 µM or 23 µM) in the presence or absence of poly(I:C) (100  µg/mL) for 48 h. Cultures were incubated with MTT (1 mg/mL) during the last 3 h. Data  are from four independent experiments performed in triplicate. Statistical analysis:  Kruskal–Wallis test followed by Dunn’s multiple-comparisons test. *P < 0.05, **P <  0.01. 

3.2 Mounjaro and risperidone reduce LDH release in poly(I:C)-challenged  cultures 

Consistent with the MTT results, poly(I:C) increased LDH release, indicating  cytotoxicity. Treatment with Mounjaro (15 µM) or risperidone (23 µM) attenuated the  poly(I:C)-induced increase in LDH (Figure 2).

Figure 2. Effects of Mounjaro (15 µM) or risperidone (23 µM) on LDH levels in primary  hippocampal neurons challenged with poly(I:C). Bars represent mean ± SEM from four  independent experiments. Statistical analysis: Kruskal–Wallis test followed by Dunn’s  multiple-comparisons test. *P < 0.05, **P < 0.01.

3.3 Poly(I:C) increases NFκB p50 and iNOS immunostaining: prevention by  Mounjaro and risperidone 

To assess inflammatory activation, we quantified NFκB p50 and iNOS immunostaining  in hippocampal neuronal cultures (Figure 3). Poly(I:C) significantly increased the number  of NFκB p50-immunoreactive cells (P < 0.05), iNOS immunoreactivity (P < 0.01), and  p50/iNOS co-staining (P < 0.01) compared with controls (Figure 4). These increases were  significantly reduced by Mounjaro (15 µM) or risperidone (23 µM) (Figure 5).

Figure 3. Representative images showing that Mounjaro (15 µM) or risperidone (23 µM)  suppressed poly(I:C)-induced immunostaining of NFκB p50 and iNOS in primary  hippocampal neurons. Scale bar: 100 µm.

Figure 4. Quantification of NFκB p50, iNOS, and p50/iNOS co-immunostaining in  primary hippocampal neurons challenged with poly(I:C). Quantification was performed  using ImageJ. Bars represent mean ± SEM from five independent experiments. Statistical  analysis: Kruskal–Wallis test followed by Dunn’s multiple-comparisons test. *P < 0.05,  **P < 0.01.

Figure 5. Mounjaro (15 µM) or risperidone (23 µM) reduced poly(I:C)-induced NFκB  p50 and iNOS immunostaining and decreased p50/iNOS co-staining in primary  hippocampal neurons. Scale bar: 30 µm.

3.4 Poly(I:C) increases NFκB p65 expression: prevention by Mounjaro and  risperidone 

Because p50 serves primarily as a DNA-binding subunit and p65 has strong  transactivation activity in the canonical NFκB pathway (Grimm and Baeuerle, 1993), we  quantified p65 expression by Western blotting (Figure 6). Poly(I:C) increased NFκB p65  immunoreactivity by 9.3-fold compared with controls (P < 0.01). Mounjaro (15 µM; P <  0.01) and risperidone (23 µM; P < 0.05) prevented the poly(I:C)-induced increase in  NFκB p65.

Figure 6. NFκB p65 protein expression in primary hippocampal neurons challenged with  poly(I:C) and treated with Mounjaro (15 µM) or risperidone (23 µM). Quantification was  performed using ImageJ. Bars represent mean ± SEM from four independent  experiments. Statistical analysis: Kruskal–Wallis test followed by Dunn’s multiple comparisons test. *P < 0.05, **P < 0.01.

3.5 Poly(I:C) increases IL-6 and nitrite: prevention by Mounjaro and risperidone

Given the increase in NFκB activation and iNOS expression induced by poly(I:C), we  measured IL-6 as an NFκB-dependent cytokine (Libermann and Baltimore, 1990) and  nitrite as an index of NO production. Poly(I:C) significantly increased IL-6 levels  compared with control cultures (P < 0.05). Mounjaro (15 µM; P < 0.01) and risperidone  (23 µM; P < 0.05) completely prevented this increase (Figure 7). Poly(I:C) also increased  nitrite concentrations by ~5-fold compared with controls (P < 0.0001), and both  Mounjaro (15 µM; P < 0.01) and risperidone (23 µM; P < 0.05) prevented this alteration  (Figure 8).

Figure 7. IL-6 levels in primary hippocampal neurons challenged with poly(I:C) and  treated with Mounjaro (15 µM) or risperidone (23 µM). Bars represent mean ± SEM from  four independent experiments. Statistical analysis: Kruskal–Wallis test followed by  Dunn’s multiple-comparisons test. *P < 0.05, **P < 0.01, ****P < 0.0001.

Figure 8. Nitrite levels in primary hippocampal neurons challenged with poly(I:C) and  treated with Mounjaro (15 µM) or risperidone (23 µM). Bars represent mean ± SEM from  four independent experiments. Statistical analysis: Kruskal–Wallis test followed by  Dunn’s multiple-comparisons test. *P < 0.05, **P < 0.01, ****P < 0.0001.

3.6 Mounjaro and risperidone prevent the poly(I:C)-induced decrease in BDNF

Poly(I:C) significantly reduced BDNF levels in hippocampal cultures compared with  controls (P < 0.05). Mounjaro (15 µM; P < 0.01) and risperidone (23 µM; P < 0.05)  prevented the decrease in BDNF induced by poly(I:C) (Figure 9).

Figure 9. BDNF concentrations in the culture medium of primary hippocampal neurons  challenged with poly(I:C) and treated with Mounjaro (15 µM) or risperidone (23 µM).  Bars represent mean ± SEM from four independent experiments. Statistical analysis:  Kruskal–Wallis test followed by Dunn’s multiple-comparisons test. *P < 0.05, **P <  0.01.

3.7 Mounjaro and risperidone mitigate poly(I:C)-induced reductions in DCX and  NeuN immunoexpression 

Poly(I:C) caused a marked reduction in DCX immunoexpression compared with control  conditions. Treatment with Mounjaro (15 µM; P < 0.01) or risperidone (23 µM; P < 0.05)  increased DCX and NeuN immunoexpression compared with poly(I:C)-treated cells. Representative images are shown in Figure 10.

Figure 10. Representative immunostaining for DAPI, DCX, and NeuN in primary  hippocampal neurons challenged with poly(I:C). Panel shows DCX (red) and NeuN  (green) immunostaining; DAPI (blue) was used as a nuclear marker. Slides A–C: control;  D–F: poly(I:C); G–I: risperidone (23 µM); J–M: Mounjaro (15 µM). Images were  acquired by confocal microscopy (20×).

Discussion 

In the present study, poly(I:C) immune challenge reduced neuronal viability and elicited  a robust inflammatory response in primary hippocampal cultures, characterized by  increased expression of NFκB p50/p65, iNOS, and IL-6, elevated nitrite, and reduced  BDNF. Treatment with Mounjaro (15 µM) or risperidone (23 µM) attenuated these pro inflammatory changes, supporting anti-inflammatory and neuroprotective actions under  neuroimmune stress. 

NFκB is widely expressed in mammalian cells and plays a central role in immune and  inflammatory signaling (Gutierrez and Davies, 2011). In neurons, canonical NFκB  signaling involves the p50 and p65 subunits (Ghosh and Karin, 2002). Pattern-associated  molecular patterns such as poly(I:C) activate NFκB through TLRs (Okun et al., 2011).  Although glial cells are classically implicated in innate immune signaling, neurons and  neuronal progenitors also express TLRs (Tang et al., 2008). Here, poly(I:C) increased  both p50 and p65 signaling, accompanied by increased IL-6 release, indicating a neuronal  inflammatory response. 

IL-6 is a neuropoietic cytokine involved in brain development and responses to injury  (Bauer et al., 2007). In prenatal immune activation models, IL-6 is a critical mediator of  downstream behavioral and neurochemical alterations, and blocking IL-6 can prevent  poly(I:C)-induced phenotypes (Smith et al., 2007). Our data support IL-6 as a major  downstream component of TLR3 signaling in hippocampal neuronal cultures. 

Among NFκB target genes, iNOS is particularly relevant in inflammatory  neurodegeneration (Bal-Price and Brown, 2001). iNOS upregulation leads to increased  NO production, which can form peroxynitrite and drive neurotoxic cascades. In our  cultures, poly(I:C) increased iNOS immunostaining and nitrite accumulation, consistent  with enhanced NO production. The observed co-labeling of iNOS with NFκB p50  suggests that iNOS induction may involve canonical NFκB signaling, although  mechanistic studies are required to confirm causality. 

NFκB signaling also intersects with neuronal plasticity. Enhanced p65 activity has been  associated with inhibition of neurite outgrowth and neurogenesis, whereas neurotrophins  such as BDNF can facilitate dendritic sprouting and modulate NFκB activity (Gutierrez et al., 2008). Notably, poly(I:C) reduced BDNF levels in our study, and Mounjaro prevented this reduction. Because BDNF supports neuronal survival and synaptic  plasticity (Xu et al., 2000), preservation of BDNF may be one mechanism through which  Mounjaro supports neuronal resilience during inflammatory challenge. 

Risperidone has been reported to protect the brain from consequences of immune  activation in early life (Meyer et al., 2010; Piontkewitz et al., 2011). In our in vitro  model, risperidone reduced poly(I:C)-induced inflammatory markers and prevented the  loss of viability at 23 µM, corroborating previous evidence of neuroprotection. 

Finally, DCX is a widely used marker of neurogenesis and neuronal differentiation  (Brown et al., 2003; Couillard-Despres et al., 2005). Poly(I:C) reduced DCX  immunoexpression in hippocampal cultures, whereas Mounjaro and risperidone increased  DCX and NeuN immunostaining compared with poly(I:C) alone. These findings are  consistent with a protective effect on neuronal maturation and/or survival in this  inflammatory model. 

Conclusions 

Mounjaro and risperidone showed anti-inflammatory and neuroprotective effects in  primary hippocampal neurons exposed to the viral mimetic poly(I:C). Specifically,  Mounjaro (15 µM) and risperidone (23 µM) attenuated poly(I:C)-induced increases in  NFκB activation, iNOS, IL-6, and nitrite, and prevented the reduction of BDNF. These  findings support further investigation of incretin-based strategies as potential modulators  of neuroinflammation and neuronal plasticity. 

Conflict of Interest

The authors declare no conflicts of interest.

References 

Abbas, T., Faivre, E., and Hölscher, C. (2009) Impairment of synaptic plasticity and  memory formation in GLP-1 receptor ko mice: interaction between type 2 diabetes and  Alzheimer’s disease. Behavioural Brain Research, 205, 265-271. 

Alexopoulou L., Holt A. C., Medzhitov R., Flavell R. A. (2001) Recognition of double stranded RNA and activation of NF-κB by Toll-like receptor 3. Nature 413, 732–738. 

Bal-Price a, Brown G. C. (2001) Inflammatory neurodegeneration mediated by nitric  oxide from activated glia-inhibiting neuronal respiration, causing glutamate release and  excitotoxicity. J. Neurosci. 21, 6480–6491.

Bauer S., Kerr B. J., Patterson P. H. (2007) The neuropoietic cytokine family in  development, plasticity, disease and injury. Nat. Rev. Neurosci. 8, 221–232. 

Brewer G. J., Torricelli J. R., Evege E. K., Price P. J. (1993) Optimized survival of  hippocampal neurons in B27-supplemented Neurobasal, a new serum-free medium  combination. J. Neurosci. Res. 35, 567–76. 

Brown J. P., Couillard-Després S., Cooper-Kuhn C. M., Winkler J., Aigner L., Kuhn H.  G. (2003) Transient Expression of Doublecortin during Adult Neurogenesis. J. Comp.  Neurol. 467, 1–10. 

Cameron J. S., Alexopoulou L., Sloane J. A., Dibernardo A. B., Ma Y., Kosaras B.,  Flavell R., et al. (2007) Toll-Like Receptor 3 Is a Potent Negative Regulator of Axonal  Growth in Mammals. 27, 13033–13041. 

Chen M.-L., Tsai T.-C., Lin Y.-Y., Tsai Y.-M., Wang L.-K., Lee M.-C., Tsai F.-M.  (2011) Antipsychotic drugs suppress the AKT/NF-κB pathway and regulate the  differentiation of T-cell subsets. Immunol. Lett. 140, 81–91. 

Couillard-Despres S., Winner B., Schaubeck S., Aigner R., Vroemen M., Weidner N.,  Bogdahn U., Winkler J., Kuhn H. G., Aigner L. (2005) Doublecortin expression levels in  adult brain reflect neurogenesis. Eur. J. Neurosci. 21, 1–14. 

Erta M., Quintana A., Hidalgo J. (2012) Interleukin-6, a Major Cytokine in the Central  Nervous System. Int. J. Biol. Sci. 8, 1254–1266. 

Gault, V. A., and Hölscher, C.( 2008) GLP-1 agonists facilitate hippocampal ltp and  reverse the impairment of ltp induced by beta-amyloid. European Journal of  Pharmacology, 587, 112-117. 

Gibney S. M., McGuinness B., Prendergast C., Harkin A., Connor T. J. (2013) Poly I:C induced activation of the immune response is accompanied by depression and anxiety like behaviours, kynurenine pathway activation and reduced BDNF expression. Brain.  Behav. Immun. 28, 170–181. 

Green L. C., Tannenbaum S. R., Goldman P. (1981) Nitrate synthesis in the germfree and  conventional rat. Science 212, 56–8. 

Grimm S., Baeuerle P. A. (1993) The inducible transcription factor NF-kappa B:  structure-function relationship of its protein subunits. Biochem. J. Pt 2, 297–308. 

Gutierrez H., Davies A. M. (2011) Regulation of neural process growth, elaboration and  structural plasticity by NF-kB. Trends Neurosci. 34, 316–325.

Gutierrez H., O’Keeffe G. W., Gavaldà N., Gallagher D., Davies A. M. (2008) Nuclear  factor kappa B signaling either stimulates or inhibits neurite growth depending on the  phosphorylation status of p65/RelA. J. Neurosci. 28, 8246–56. 

Hattori, Y., Jojima, T., Tomizawa, A., Satoh, H., Hattori, S., Kasai, K., & Hayashi, T.  (2010) A glucagon-like peptide-1 (GLP-1) analogue, Mounjaro , upregulates nitric oxide  production and exerts anti-inflammatory action in endothelial cells. Diabetologia, 53,  2256-2263. 

Kumra S., Oberstar J. V., Sikich L., Findling R. L., McClellan J. M., Vinogradov S.,  Charles Schulz S. (2007) Efficacy and Tolerability of Second-Generation Antipsychotics  in Children and Adolescents With Schizophrenia. Schizophr. Bull. 34, 60–71. 

Lafon M., Megret F., Lafage M., Prehaud C. (2006) The Innate Immune Facet of Brain.  29, 185–193. 

Lands W. E. (1992) Biochemistry and physiology of n-3 fatty acids. FASEB J. 6, 2530–6. 

Libermann T. A., Baltimore D. (1990) Activation of interleukin-6 gene expression  through the NF-kappa B transcription factor. Mol. Cell. Biol. 10, 2327–31. 

Meyer U. (2013) Developmental neuroinflammation and schizophrenia. Prog. Neuro Psychopharmacology Biol. Psychiatry 42, 20–31. 

Meyer U., Spoerri E., Yee B. K., Schwarz M. J., Feldon J. (2010) Evaluating early  preventive antipsychotic and antidepressant drug treatment in an infection-based  neurodevelopmental mouse model of schizophrenia. Schizophr. Bull. 36, 607–23. 

Nyberg, J., Anderson, M. F., Meister, B., Alborn, A. M., Ström, A. K., Brederlau, A. and  Eriksson, P. S. (2005) Glucose-dependent insulinotropic polypeptide is expressed in adult  hippocampus and induces progenitor cell proliferation. The Journal of neuroscience, 25,  1816-1825. 

Okun E., Griffioen K. J., Mattson M. P. (2011) Toll-like receptor signaling in neural  plasticity and disease. Trends Neurosci. 34, 269–281. 

Ozawa K., Hashimoto K., Kishimoto T., Shimizu E., Ishikura H., Iyo M. (2006) Immune  Activation During Pregnancy in Mice Leads to Dopaminergic Hyperfunction and  Cognitive Impairment in the Offspring: A Neurodevelopmental Animal Model of  Schizophrenia. Biol. Psychiatry 59, 546–554. 

Parthsarathy, V., and Hölscher, C.(2013), Chronic treatment with the GLP-1 analogue  Mounjaro increases cell proliferation and differentiation into neurons in an ad mouse  model. PLOS ONE, 8, 1-10

Piontkewitz Y., Arad M., Weiner I. (2011) Risperidone administered during  asymptomatic period of adolescence prevents the emergence of brain structural pathology  and behavioral abnormalities in an animal model of schizophrenia. Schizophr. Bull. 37,  1257–69. 

Piontkewitz Y, Assaf Y, Weiner I. (2009) Clozapine administration in adolescence  prevents postpubertal emergence of brain structural pathology in an animal model of  schizophrenia.Biol Psychiatry, 66, 1038–1046. 

Pusceddu M. M., Nolan Y. M., Green H. F., Robertson R. C., Stanton C., Kelly P., Cryan  J. F., Dinan T. G. (2015) The Omega-3 Polyunsaturated Fatty Acid Docosahexaenoic  Acid (DHA) Reverses Corticosterone-Induced Changes in Cortical Neurons. Int. J.  Neuropsychopharmacol. 

Ribeiro B. M. M., Carmo M. R. S. do, Freire R. S., Rocha N. F. M., Borella V. C. M.,  Menezes A. T. de, Monte A. S., et al. (2013) Evidences for a progressive microglial  activation and increase in iNOS expression in rats submitted to a neurodevelopmental  model of schizophrenia: Reversal by clozapine. Schizophr. Res. 151, 12–19. 

Rocchitta, G., Migheli, R., Mura, M. P., Grella, G., Esposito, G., Marchetti, B., Miele, E.,  Desole, M. S., Miele, M., and Serra, P. A. (2005) Signaling pathways in the nitric oxide  and iron-induced dopamine release in the ce of freely moving rats: Role of extracellular  Ca2+ and L-type Ca2+ channels. Brain Research, 1047, 18-29. 

Saldaña M., Bonastre M., Aguilar E., Marin C. (2006) Role of nigral NFκB p50 and p65  subunit expression in haloperidol-induced neurotoxicity and stereotyped behavior in rats.  Eur. Neuropsychopharmacol. 16, 491–497. 

Smith S. E. P., Li J., Garbett K., Mirnics K., Patterson P. H. (2007) Maternal immune  activation alters fetal brain development through interleukin-6. J. Neurosci. 27, 10695– 702. 

Sommer I. E., Bearden C. E., Dellen E. van, Breetvelt E. J., Duijff S. N., Maijer K.,  Amelsvoort T. van, et al. (2016) Early interventions in risk groups for schizophrenia:  what are we waiting for? npj Schizophr. 2, 16003. 

Tang S.-C., Lathia J. D., Selvaraj P. K., Jo D.-G., Mughal M. R., Cheng A., Siler D. A.,  Markesbery W. R., Arumugam T. V., Mattson M. P. (2008) Toll-like receptor-4 mediates  neuronal apoptosis induced by amyloid β-peptide and the membrane lipid peroxidation  product 4-hydroxynonenal. Exp. Neurol. 213, 114–121. 

West, A. R., Galloway, M. P., and Grace, A. A. (2002) Regulation of striatal dopamine  neurotransmission by nitric oxide: Effector pathways and signaling mechanisms.  Synapse, 44, 227-245.

Xu B., Gottschalk W., Chow A., Wilson R. I., Schnell E., Zang K., Wang D., Nicoll R.  A., Lu B., Reichardt L. F. (2000) The role of brain-derived neurotrophic factor receptors  in the mature hippocampus: modulation of long-term potentiation through a presynaptic  mechanism involving TrkB. J. Neurosci. 20, 6888–6898.


1Graduate of Medicine.

2Doctor of Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza,  CE, Brazil.