Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
2.
Front Immunol ; 12: 709861, 2021.
Article in English | MEDLINE | ID: mdl-34475873

ABSTRACT

BACKGROUND: Immune hyperactivity is an important contributing factor to the morbidity and mortality of COVID-19 infection. Nasal administration of anti-CD3 monoclonal antibody downregulates hyperactive immune responses in animal models of autoimmunity through its immunomodulatory properties. We performed a randomized pilot study of fully-human nasal anti-CD3 (Foralumab) in patients with mild to moderate COVID-19 to determine if its immunomodulatory properties had ameliorating effects on disease. METHODS: Thirty-nine outpatients with mild to moderate COVID-19 were recruited at Santa Casa de Misericordia de Santos in Sao Paulo State, Brazil. Patients were randomized to three cohorts: 1) Control, no Foralumab (n=16); 2) Nasal Foralumab (100ug/day) given for 10 consecutive days with 6 mg dexamethasone given on days 1-3 (n=11); and 3) Nasal Foralumab alone (100ug/day) given for 10 consecutive days (n=12). Patients continued standard of care medication. RESULTS: We observed reduction of serum IL-6 and C-reactive protein in Foralumab alone vs. untreated or Foralumab/Dexa treated patients. More rapid clearance of lung infiltrates as measured by chest CT was observed in Foralumab and Foralumab/Dexa treated subjects vs. those that did not receive Foralumab. Foralumab treatment was well-tolerated with no severe adverse events. CONCLUSIONS: This pilot study suggests that nasal Foralumab is well tolerated and may be of benefit in treatment of immune hyperactivity and lung involvement in COVID-19 disease and that further studies are warranted.


Subject(s)
Antibodies, Monoclonal/therapeutic use , COVID-19/immunology , COVID-19/prevention & control , Pneumonia/therapy , Administration, Intranasal , Adolescent , Adult , Antibodies, Monoclonal/administration & dosage , Biomarkers , C-Reactive Protein/analysis , COVID-19/physiopathology , COVID-19/therapy , Cohort Studies , Female , Humans , Immunity/drug effects , Interleukin-6/blood , Lung/drug effects , Lung/immunology , Lung/pathology , Male , Middle Aged , Outpatients/statistics & numerical data , Pilot Projects , Pneumonia/prevention & control , Young Adult
3.
World J Gastroenterol ; 24(17): 1888-1900, 2018 May 07.
Article in English | MEDLINE | ID: mdl-29740204

ABSTRACT

AIM: To investigate the effects of plecanatide and dolcanatide on maintenance of paracellular permeability, integrity of tight junctions and on suppression of visceral hypersensitivity. METHODS: Transport of fluorescein isothiocyanate (FITC)-dextran was measured to assess permeability across cell monolayers and rat colon tissues. Effects of plecanatide and dolcanatide on the integrity of tight junctions in Caco-2 and T84 monolayers and on the expression and localization of occludin and zonula occludens-1 (ZO-1) were examined by immunofluorescence microscopy. Anti-nociceptive activity of these agonists was evaluated in trinitrobenzene sulfonic acid (TNBS)-induced inflammatory as well as in non-inflammatory partial restraint stress (PRS) rat models. Statistical significance between the treatment groups in the permeability studies were evaluated using unpaired t-tests. RESULTS: Treatment of T84 and Caco-2 monolayers with lipopolysaccharide (LPS) rapidly increased permeability, which was effectively suppressed when monolayers were also treated with plecanatide or dolcanatide. Similarly, when T84 and Caco-2 monolayers were treated with LPS, cell surface localization of tight junction proteins occludin and ZO-1 was severely disrupted. When cell monolayers were treated with LPS in the presence of plecanatide or dolcanatide, occludin and ZO-1 were localized at the cell surface of adjoining cells, similar to that observed for vehicle treated cells. Treatment of cell monolayers with plecanatide or dolcanatide without LPS did not alter permeability, integrity of tight junctions and cell surface localization of either of the tight junction proteins. In rat visceral hypersensitivity models, both agonists suppressed the TNBS-induced increase in abdominal contractions in response to colorectal distension without affecting the colonic wall elasticity, and both agonists also reduced colonic hypersensitivity in the PRS model. CONCLUSION: Our results suggest that activation of GC-C signaling might be involved in maintenance of barrier function, possibly through regulating normal localization of tight junction proteins. Consistent with these findings, plecanatide and dolcanatide showed potent anti-nociceptive activity in rat visceral hypersensitivity models. These results imply that activation of GC-C signaling may be an attractive therapeutic approach to treat functional constipation disorders and inflammatory gastrointestinal conditions.


Subject(s)
Constipation/drug therapy , Guanylyl Cyclase C Agonists/pharmacology , Irritable Bowel Syndrome/drug therapy , Receptors, Enterotoxin/metabolism , Visceral Pain/drug therapy , Administration, Oral , Animals , Caco-2 Cells , Colon/cytology , Colon/drug effects , Colon/pathology , Constipation/pathology , Dextrans/pharmacokinetics , Female , Fluorescein-5-isothiocyanate/analogs & derivatives , Fluorescein-5-isothiocyanate/pharmacokinetics , Guanylyl Cyclase C Agonists/therapeutic use , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Irritable Bowel Syndrome/etiology , Irritable Bowel Syndrome/pathology , Lipopolysaccharides/pharmacology , Male , Natriuretic Peptides/pharmacology , Natriuretic Peptides/therapeutic use , Nociception/drug effects , Peptides/pharmacology , Peptides/therapeutic use , Permeability/drug effects , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Tight Junctions/drug effects , Tight Junctions/metabolism , Trinitrobenzenesulfonic Acid/toxicity , Visceral Pain/chemically induced , Visceral Pain/pathology
4.
World J Gastrointest Pharmacol Ther ; 8(1): 47-59, 2017 Feb 06.
Article in English | MEDLINE | ID: mdl-28217374

ABSTRACT

AIM: To evaluate the effect of orally administered plecanatide on colorectal dysplasia in Apc+/Min-FCCC mice with dextran sodium sulfate (DSS)-induced inflammation. METHODS: Inflammation driven colorectal carcinogenesis was induced in Apc+/Min-FCCC mice by administering DSS in their drinking water. Mice were fed a diet supplemented with plecanatide (0-20 ppm) and its effect on the multiplicity of histopathologically confirmed polypoid, flat and indeterminate dysplasia was evaluated. Plecanatide-mediated activation of guanylate cyclase-C (GC-C) signaling was assessed in colon tissues by measuring cyclic guanosine monophosphate (cGMP) by ELISA, protein kinase G-II and vasodilator stimulated phosphoprotein by immunoblotting. Ki-67, c-myc and cyclin D1 were used as markers of proliferation. Cellular levels and localization of ß-catenin in colon tissues were assessed by immunoblotting and immunohistochemistry, respectively. Uroguanylin (UG) and GC-C transcript levels were measured by quantitative reverse transcription polymerase chain reaction (RT-PCR). A mouse cytokine array panel was used to detect cytokines in the supernatant of colon explant cultures. RESULTS: Oral treatment of Apc+/MinFCCC mice with plecanatide produced a statistically significant reduction in the formation of inflammation-driven polypoid, flat and indeterminate dysplasias. This anti-carcinogenic activity of plecanatide was accompanied by activation of cGMP/GC-C signaling mediated inhibition of Wnt/ß-catenin signaling and reduced proliferation. Plecanatide also decreased secretion of pro-inflammatory cytokines (IL-6, IL1 TNF), chemokines (MIP-1, IP-10) and growth factors (GCSF and GMCSF) from colon explants derived from mice with acute DSS-induced inflammation. The effect of plecanatide-mediated inhibition of inflammation/dysplasia on endogenous expression of UG and GC-C transcripts was measured in intestinal tissues. Although GC-C expression was not altered appreciably, a statistically significant increase in the level of UG transcripts was detected in the proximal small intestine and colon, potentially due to a reduction in intestinal inflammation and/or neoplasia. Taken together, these results suggest that reductions in endogenous UG, accompanied by dysregulation in GC-C signaling, may be an early event in inflammation-promoted colorectal neoplasia; an event that can potentially be ameliorated by prophylactic intervention with plecanatide. CONCLUSION: This study provides the first evidence that orally administered plecanatide reduces the multiplicity of inflammation-driven colonic dysplasia in mice, demonstrating the utility for developing GC-C agonists as chemopreventive agents.

5.
World J Gastrointest Pharmacol Ther ; 6(4): 213-22, 2015 Nov 06.
Article in English | MEDLINE | ID: mdl-26558155

ABSTRACT

AIM: To evaluate the effect of orally administered plecanatide or dolcanatide, analogs of uroguanylin, on amelioration of colitis in murine models. METHODS: The cyclic guanosine monophosphate (cGMP) stimulatory potency of plecanatide and dolcanatide was measured using a human colon carcinoma T84 cell-based assay. For animal studies all test agents were formulated in phosphate buffered saline. Sulfasalazine or 5-amino salicylic acid (5-ASA) served as positive controls. Effect of oral treatment with test agents on amelioration of acute colitis induced either by dextran sulfate sodium (DSS) in drinking water or by rectal instillation of trinitrobenzene sulfonic (TNBS) acid, was examined in BALB/c and/or BDF1 mice. Additionally, the effect of orally administered plecanatide on the spontaneous colitis in T-cell receptor alpha knockout (TCRα(-/-)) mice was also examined. Amelioration of colitis was assessed by monitoring severity of colitis, disease activity index and by histopathology. Frozen colon tissues were used to measure myeloperoxidase activity. RESULTS: Plecanatide and dolcanatide are structurally related analogs of uroguanylin, which is an endogenous ligand of guanylate cyclase-C (GC-C). As expected from the agonists of GC-C, both plecanatide and dolcanatide exhibited potent cGMP-stimulatory activity in T84 cells. Once-daily treatment by oral gavage with either of these analogs (0.05-0.5 mg/kg) ameliorated colitis in both DSS and TNBS-induced models of acute colitis, as assessed by body weight, reduction in colitis severity (P < 0.05) and disease activity index (P < 0.05). Amelioration of colitis by either of the drug candidates was comparable to that achieved by orally administered sulfasalazine or 5-ASA. Plecanatide also effectively ameliorated colitis in TCRα(-/-) mice, a model of spontaneous colitis. As dolcanatide exhibited higher resistance to proteolysis in simulated gastric and intestinal juices, it was selected for further studies. CONCLUSION: This is the first-ever study reporting the therapeutic utility of GC-C agonists as a new class of orally delivered and mucosally active drug candidates for the treatment of inflammatory bowel diseases.

6.
Obesity (Silver Spring) ; 19(10): 1923-34, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21799481

ABSTRACT

Cannabinoid receptor CB1 is expressed abundantly in the brain and presumably in the peripheral tissues responsible for energy metabolism. It is unclear if the antiobesity effects of rimonabant, a CB1 antagonist, are mediated through the central or the peripheral CB1 receptors. To address this question, we generated transgenic mice with central nervous system (CNS)-specific knockdown (KD) of CB1, by expressing an artificial microRNA (AMIR) under the control of the neuronal Thy1.2 promoter. In the mutant mice, CB1 expression was reduced in the brain and spinal cord, whereas no change was observed in the superior cervical ganglia (SCG), sympathetic trunk, enteric nervous system, and pancreatic ganglia. In contrast to the neuronal tissues, CB1 was undetectable in the brown adipose tissue (BAT) or the liver. Consistent with the selective loss of central CB1, agonist-induced hypothermia was attenuated in the mutant mice, but the agonist-induced delay of gastrointestinal transit (GIT), a primarily peripheral nervous system-mediated effect, was not. Compared to wild-type (WT) littermates, the mutant mice displayed reduced body weight (BW), adiposity, and feeding efficiency, and when fed a high-fat diet (HFD), showed decreased plasma insulin, leptin, cholesterol, and triglyceride levels, and elevated adiponectin levels. Furthermore, the therapeutic effects of rimonabant on food intake (FI), BW, and serum parameters were markedly reduced and correlated with the degree of CB1 KD. Thus, KD of CB1 in the CNS recapitulates the metabolic phenotype of CB1 knockout (KO) mice and diminishes rimonabant's efficacy, indicating that blockade of central CB1 is required for rimonabant's antiobesity actions.


Subject(s)
Anti-Obesity Agents/pharmacology , Body Weight/drug effects , Central Nervous System/metabolism , Energy Intake/drug effects , Obesity/metabolism , Piperidines/pharmacology , Pyrazoles/pharmacology , Receptor, Cannabinoid, CB1/metabolism , Adiponectin/blood , Adiposity/drug effects , Adiposity/genetics , Animals , Anti-Obesity Agents/therapeutic use , Biomarkers/blood , Body Weight/genetics , Central Nervous System/drug effects , Cholesterol/blood , Diet, High-Fat/adverse effects , Energy Intake/genetics , Gastrointestinal Transit/physiology , Hypothermia/prevention & control , Insulin/blood , Leptin/blood , Mice , Mice, Knockout , Mice, Transgenic , MicroRNAs , Mutation , Obesity/drug therapy , Obesity/genetics , Peripheral Nervous System/drug effects , Peripheral Nervous System/metabolism , Phenotype , Piperidines/therapeutic use , Promoter Regions, Genetic , Pyrazoles/therapeutic use , Receptor, Cannabinoid, CB1/genetics , Rimonabant , Triglycerides/blood
7.
Mol Cell Endocrinol ; 325(1-2): 18-25, 2010 Aug 30.
Article in English | MEDLINE | ID: mdl-20416356

ABSTRACT

The free fatty acid (FFA) receptor GPR40, expressed by pancreatic beta-cells, may be responsible for insulin release following beta(3) adrenoceptor (Adrb3) activation. To test this hypothesis, we first studied the effects of Adrb3 agonists SR58611A and CL316,243 in GPR40 knockout (GPR40(-/-)) mice. Both drugs increased blood FFA levels in wild-type (GPR40(+/+)) and GPR40(-/-) mice, indicating that lipolysis is not GPR40-dependent. However, the magnitude of the insulin response after agonist treatment was decreased by approximately 50% in GPR40(-/-) mice. Analysis of the time-course revealed that the change in FFAs (5-10 min post-treatment) in response to SR58611A preceded insulin secretion (10-15 min post-treatment). While reduced by agonist treatment, glucose levels in GPR40(-/-) mice remained significantly higher than in GPR40(+/+) mice. Energy expenditure, food intake, or body weight was not affected in GPR40(-/-) mice, whereas SR58611A increased energy metabolism. Furthermore, CL316,243 did not potentiate glucose-stimulated insulin secretion in isolated mouse islets or activate a cAMP reporter in transgenic mice. Our data indicate that insulin secretion, a secondary event following stimulation of Adrb3 receptors, is partially mediated by GPR40 and suggest that GPR40 is integral to the anti-diabetes effects of Adrb3 agonists.


Subject(s)
Adrenergic beta-3 Receptor Agonists , Adrenergic beta-Agonists/pharmacology , Insulin/metabolism , Receptors, G-Protein-Coupled/physiology , Animals , Body Weight/drug effects , Body Weight/genetics , Dioxoles/pharmacology , Dose-Response Relationship, Drug , Eating/drug effects , Eating/genetics , Energy Metabolism/drug effects , Energy Metabolism/genetics , Hypoglycemic Agents/pharmacology , Insulin Secretion , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Adrenergic, beta-3/metabolism , Receptors, G-Protein-Coupled/genetics , Tetrahydronaphthalenes/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL