9 results on '"Guzmán-Mejía, Fabiola"'
Search Results
2. Moderate Aerobic Exercise Induces Homeostatic IgA Generation in Senile Mice.
- Author
-
Hernández-Urbán, Angel J., Drago-Serrano, Maria-Elisa, Reséndiz-Albor, Aldo A., Sierra-Ramírez, José A., Guzmán-Mejía, Fabiola, Oros-Pantoja, Rigoberto, and Godínez-Victoria, Marycarmen
- Subjects
PLASMA cells ,IMMUNOGLOBULIN class switching ,THYMIC stromal lymphopoietin ,EPITHELIAL cells ,NITRIC-oxide synthases ,B cells - Abstract
A T-cell-independent (TI) pathway activated by microbiota results in the generation of low-affinity homeostatic IgA with a critical role in intestinal homeostasis. Moderate aerobic exercise (MAE) provides a beneficial impact on intestinal immunity, but the action of MAE on TI-IgA generation under senescence conditions is unknown. This study aimed to determine the effects of long-term MAE on TI-IgA production in young (3 month old) BALB/c mice exercised until adulthood (6 months) or aging (24 months). Lamina propria (LP) from the small intestine was obtained to determine B cell and plasma cell sub-populations by flow cytometry and molecular factors related to class switch recombination [Thymic Stromal Lymphopoietin (TSLP), A Proliferation-Inducing Ligand (APRIL), B Cell Activating Factor (BAFF), inducible nitric oxide synthase (iNOS), and retinal dehydrogenase (RDH)] and the synthesis of IgA [α-chain, interleukin (IL)-6, IL-21, and Growth Factor-β (TGF-β)]; and epithelial cells evaluated IgA transitosis [polymeric immunoglobulin receptor (pIgR), tumor necrosis factor-α (TNF-α), interferon-γ (IFN-γ), IL-4] by the RT-qPCR technique. The results were compared with data obtained from sedentary age-matched mice. Statistical analysis was computed with ANOVA, and p < 0.05 was considered to be a statistically significant difference. Under senescence conditions, MAE promoted the B cell and IgA+ B cells and APRIL, which may improve the intestinal response and ameliorate the inflammatory environment associated presumably with the downmodulation of pro-inflammatory mediators involved in the upmodulation of pIgR expression. Data suggested that MAE improved IgA and downmodulate the cytokine pro-inflammatory expression favoring homeostatic conditions in aging. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
3. Effect of chronic stress on gel-forming mucins in the small intestine of BALB/c mice.
- Author
-
Gutiérrez-Galicia, Jennifer Karume, Drago-Serrano, Maria Elisa, Oros-Pantoja, Rigoberto, Godínez-Victoria, Marycarmen, and Guzmán-Mejía, Fabiola
- Abstract
Intestinal homeostasis involves the collaboration of gut barrier components, such as goblet cells and IgA-microbiota complexes, that are under the control of stress that promotes inflammatory responses addressed primarily in the colon. The aim of this study was to evaluate the effect of stress on mucins, goblet cells, and proinflammatory parameters in the proximal and distal regions of the small intestine. A group (n = 6) of female 8-week-old BALB/c mice underwent board immobilization stress (2 h per day for 4 days) and were sacrificed with isoflurane. Samples from proximal and distal small segments were collected to analyze the following: 1) goblet cells stained with periodic acid-Schiff (PAS) and with alcian blue (AB) to visualize histologically neutral and acidic mucins, respectively; 2) IgA-microbiota complexes identified by flow cytometry in intestinal lavages; and 3) MUC2, MUC5AC, and IL-18 mRNA levels in whole mucosal scrapings by reverse transcription-qPCR. Regarding the unstressed group, in the proximal region of small intestine both PAS+ and AB+ goblet cells were unchanged; however, MUC5AC and IL-18 mRNA levels were increased, and the percentage of IgA-microbiota complexes was reduced. In the distal segment, the number of PAS+ goblet cells was increased, whereas the number of AB+ goblet cells was reduced and did not affect the remaining parameters. The data suggest that stress induces inflammation in the proximal small intestine; these findings may provide an experimental reference for human diseases that may affect the proximal small intestine, such as Crohn's disease, in which stress contributes to the progression of intestinal inflammation or relapse. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
4. Outcome of stress on G protein-coupled receptors and hypoxia inducible factor-1α.
- Author
-
Arturo Reséndiz-Albor, Aldo, Maciel Arciniega-Martínez, Ivonne, Montes de Oca, Arturo Contis, Guzmán-Mejía, Fabiola, Drago-Serrano, Maria Elisa, Estrada-Jiménez, Tania, and Abarca-Rojano, Edgar
- Subjects
G protein coupled receptors ,HYPOXEMIA ,WESTERN immunoblotting ,PSYCHOLOGICAL stress ,IMMOBILIZATION stress - Abstract
Stress drives neuroendocrine signals with detrimental effects to the intestinal homeostasis. The aim of this study was to evaluate the effect of stress on intestinal hypoxia response elements, including G protein-coupled receptor 41 (GPR41), GPR43, and hypoxia inducible factor (HIF)-1α. Groups of five BALB/c mice were subjected to acute (2 h per day) and chronic (2 h per day for 4 days) stress induced by restraint, and the results were compared to those of an unstressed control group. Whole mucosal samples from the colon were collected to evaluate the expression of GPR41, GPR43 and HIF-1α using Western blot chemiluminescent analysis. Compared to the control group, in the chronic stress group the expression of GPR43 (P = 0.0092) and HIF-1α (P < 0.0001) were significantly lower and the expression of GPR41 was similar (P = 0.9184); acute stress significantly increased HIF-1α expression (P = 0.0030) and increased GPR41 expression (P = 0.0937), without affecting GPR43 (P = 0.9184). These findings offer insights into the modulation of hypoxia response elements under stress conditions and their pharmacological implications for developing drugs that mitigate the effects of stress on intestinal homeostasis. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
5. Regionalized analysis of gut barrier components in the small intestine of BALB/c mice following chronic immobilization stress.
- Author
-
Guzmán-Mejía, Fabiola, Godínez-Victoria, Marycarmen, García-Hernández, Ana Lilia, Gutierrez-Galicia, Jennifer Karume, Molotla Torres, Daniel Efrain, and Drago-Serrano, Maria Elisa
- Subjects
SMALL intestine ,IMMOBILIZATION stress ,PSYCHOLOGICAL stress ,MESSENGER RNA ,TUMOR necrosis factors - Abstract
Background. Microbiota and tight junction proteins (TJPs) are components of the gut barrier, and are considered stress targets that have deleterious effects on intestinal homeostasis. Objectives. This study aimed to evaluate the effects of chronic immobilization stress on selected small intestine homeostasis parameters. Materials and methods. Female BALB/c mice were divided into a stress group that underwent short-term immobilization for 2 h per day for 4 consecutive days, and a non-stressed control group (n = 6 per group). Proximal and distal small intestine samples were excised to assess colony-forming units per gram (CFU/g) of total bifidobacteria in selective agar plates, luminal albumin was assessed using immune-enzymatic assay, pro-inflammatory cytokines were evaluated using reverse transcription-quantitative polymerase chain reaction (RT-qPCR), and TJPs (pore-forming, claudin (Cld)-2; pore-sealing, Cld-4; ambiguous, Cld-7, -12 and -15) were assessed with RT-qPCR and western blotting. Results. Compared with the control group, the stress group had lower body weight and energy intake. In the distal region, the stressed mice had lower bifidobacteria count and messenger ribonucleic acid (mRNA) expression of Cld-2, Cld-4 and Cld-12, though they had more albumin and higher interleukin (IL)-6 mRNA expression. Within the proximal region, the stressed mice had higher mRNA expression of tumor necrosis factor alpha (TNF-α), interferon gamma (IFN-γ), IL-6, Cld-7, Cld-12, and Cld-15, along with lower levels of IL-10 and Cld-4. However, mRNA and protein expression of TJPs were discordant. Conclusions. These findings indicate divergent stress-induced outcomes in the small intestine, evidenced by the elicitation of a pro-inflammatory response and decreased anti-inflammatory response in the duodenum, and by increased albumin transudation and decreased bifidobacterial growth in the distal region. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
6. Lactoferrin as a Component of Pharmaceutical Preparations: An Experimental Focus.
- Author
-
Guzmán-Mejía, Fabiola, Godínez-Victoria, Marycarmen, Molotla-Torres, Daniel Efrain, and Drago-Serrano, Maria Elisa
- Subjects
- *
LACTOFERRIN , *DRUGS , *INFLAMMATORY bowel diseases , *CENTRAL nervous system , *DRUG delivery systems , *BLOOD-brain barrier - Abstract
Lactoferrin is an 80 kDa monomeric glycoprotein that exhibits multitask activities. Lactoferrin properties are of interest in the pharmaceutical field for the design of products with therapeutic potential, including nanoparticles and liposomes, among many others. In antimicrobial preparations, lactoferrin has been included either as a main bioactive component or as an enhancer of the activity and potency of first-line antibiotics. In some proposals based on nanoparticles, lactoferrin has been included in delivery systems to transport and protect drugs from enzymatic degradation in the intestine, favoring the bioavailability for the treatment of inflammatory bowel disease and colon cancer. Moreover, nanoparticles loaded with lactoferrin have been formulated as delivery systems to transport drugs for neurodegenerative diseases, which cannot cross the blood–brain barrier to enter the central nervous system. This manuscript is focused on pharmaceutical products either containing lactoferrin as the bioactive component or formulated with lactoferrin as the carrier considering its interaction with receptors expressed in tissues as targets of drugs delivered via parenteral or mucosal administration. We hope that this manuscript provides insights about the therapeutic possibilities of pharmaceutical Lf preparations with a sustainable approach that contributes to decreasing the resistance of antimicrobials and enhancing the bioavailability of first-line drugs for intestinal chronic inflammation and neurodegenerative diseases. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
7. Oral bovine lactoferrin modulation on fecal microbiota of mice underwent immobilization stress.
- Author
-
Efrain Molotla-Torres, Daniel, Mario Hernández-Soto, Luis, Guzmán-Mejía, Fabiola, Godínez-Victoria, Marycarmen, Elisa Drago-Serrano, Maria, and Félix Aguirre-Garrido, José
- Abstract
[Display omitted] • Bovine lactoferrin leads gut microbiota dysbiosis. • Lactoferrin up or down-modulates expansion of fecal prominent microbiome lineages. • Bovine lactoferrin modulates fecal microbiome regardless stress condition. Lactoferrin, a diferric-binding glycoprotein drives the abundance of some gut microbiota members, but its role under stress conditions is unknown thus this aspect was the aim of this contribution. Female BALB/c mice (n = 4/group), treated orally with lactoferrin for 7 days were either stressed by board-immobilization from the 4th-to-7th day of treatment (LS) or unstressed (L). Two groups provided with water and stressed (S) or unstressed (C) were included. On day 7, feces were collected for the massive sequencing of V4-V5 region of the 16S rDNA gene. Regarding C -group, the only significant changes were found in the L -group that showed an increased abundance in most taxa of the genus Bacteroides (Phylum Bacteroidetes) and Parasuterella (Phylum Proteobacteria) or decreased of Clostridium _XIVb (Phylum Firmicutes), Lactoferrin-associated microbiota changes may result from stress independent modulatory pathways. Data may provide experimental foundations for using lactoferrin as a therapeutic co-adjuvant for diseases like obesity with microbiome alterations. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
8. Lactoferrin and Its Potential Impact for the Relief of Pain: A Preclinical Approach.
- Author
-
Godínez-Chaparro, Beatriz, Guzmán-Mejía, Fabiola, and Drago-Serrano, Maria Elisa
- Subjects
- *
LACTOFERRIN , *OPIOID receptors , *ACUTE kidney failure , *PAIN management , *ANALGESIA , *CELLULAR signal transduction , *STOMACH ulcers - Abstract
Pain is one of the most disabling symptoms of several clinical conditions. Neurobiologically, it is classified as nociceptive, inflammatory, neuropathic and dysfunctional. Opioids and nonsteroidal anti-inflammatory drugs (NSAIDs) are conventionally prescribed for the treatment of pain. Long-term administration of opioids results in the loss of analgesic efficacy, leading to increased dosage, tolerance, and addiction as the main drawbacks of their use, while the adverse effects of NSAIDs include gastric ulcer formation, intestinal bleeding, acute kidney injury, and hepatotoxicity. Lactoferrin is an iron-binding, anti-inflammatory glycoprotein that displays analgesic activities associated, in part, by interacting with the low-density lipoprotein receptor-related protein (LRP), which may result in the regulation of the DAMP–TRAF6–NFκB, NO–cGMP–ATP K+-sensitive channel and opioid receptor signaling pathways. This review summarizes and discusses for the first time the analgesic effects of lactoferrin and its presumable mechanisms based on pre-clinical trials. Given its anti-nociceptive and anti-inflammatory properties, lactoferrin may be used as an adjunct to enhance the efficacy and to decrease the tolerogenic effects of canonical therapeutic drugs prescribed for pain treatment. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
9. Intestinal Homeostasis under Stress Siege.
- Author
-
Guzmán-Mejía, Fabiola, Godínez-Victoria, Marycarmen, Vega-Bautista, Alan, Pacheco-Yépez, Judith, Drago-Serrano, Maria Elisa, and Subiza, José Luis
- Subjects
- *
MUCUS , *ENTERIC nervous system , *SYMPATHETIC nervous system , *IRRITABLE colon , *HOMEOSTASIS , *HYPOTHALAMIC-pituitary-adrenal axis - Abstract
Intestinal homeostasis encompasses a complex and balanced interplay among a wide array of components that collaborate to maintain gut barrier integrity. The appropriate function of the gut barrier requires the mucus layer, a sticky cushion of mucopolysaccharides that overlays the epithelial cell surface. Mucus plays a critical anti-inflammatory role by preventing direct contact between luminal microbiota and the surface of the epithelial cell monolayer. Moreover, mucus is enriched with pivotal effectors of intestinal immunity, such as immunoglobulin A (IgA). A fragile and delicate equilibrium that supports proper barrier function can be disturbed by stress. The impact of stress upon intestinal homeostasis results from neuroendocrine mediators of the brain-gut axis (BGA), which comprises a nervous branch that includes the enteric nervous system (ENS) and the sympathetic and parasympathetic nervous systems, as well as an endocrine branch of the hypothalamic-pituitary-adrenal axis. This review is the first to discuss the experimental animal models that address the impact of stress on components of intestinal homeostasis, with special emphasis on intestinal mucus and IgA. Basic knowledge from animal models provides the foundations of pharmacologic and immunological interventions to control disturbances associated with conditions that are exacerbated by emotional stress, such as irritable bowel syndrome. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.