Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Int J Cancer ; 145(5): 1189-1199, 2019 09 01.
Article in English | MEDLINE | ID: mdl-30664811

ABSTRACT

Agonistic CD40 antibodies activate dendritic cells and can expand and activate tumor-specific T cells. Our purpose was to assess the CD40 agonistic antibody ADC-1013 in the clinical setting including intratumoral administration since preclinical studies have indicated that intratumoral is better than intravenous administration. A Phase I, open label, multicenter study was conducted in patients with advanced solid tumors who had received established treatments. A modified 3 + 3 dose-escalation was applied (every other week dosing). Twenty-three patients were treated with ADC-1013 intratumorally (dosing from 22.5 µg/kg up to 400 µg/kg) or intravenously (dosing at 75 µg/kg). The pharmacodynamic effects observed in the patients were further verified in an hCD40tg mouse model. Adverse events were mostly Common Terminology Criteria for Adverse Events (CTCAE) Grades 1 or 2 and transient. The serum concentration ADC-1013 and cytokine release (MCP-1, TNFα and IL-6) were more pronounced in patients receiving injections in deep metastases compared to patients receiving injections in superficial metastases. Treatment with ADC-1013 resulted in a marked decrease in B cell levels in peripheral blood after 24 h while remaining B cells significantly increased their expression of the cell surface activation marker CD86. Activation of antigen-presenting cells and subsequent activation of T cells were demonstrated in hCD40tg mice. Moreover, ADC-1013 treatment in this mouse model acted synergistically with a PD-1 inhibitor. The results from the first-in-human study of ADC-1013 indicate that intratumoral administration of ADC-1013 into superficial lesions is well tolerated at clinically relevant doses and associated with pharmacodynamic responses.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Neoplasms/drug therapy , Adult , Aged , Animals , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antibodies, Monoclonal, Humanized/pharmacology , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Cell Line, Tumor , Dose-Response Relationship, Drug , Female , Humans , Infusions, Intralesional , Infusions, Intravenous , Macaca fascicularis , Male , Melanoma, Experimental/drug therapy , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Middle Aged , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/pathology , Young Adult
2.
Cancer Immunol Immunother ; 66(1): 1-7, 2017 01.
Article in English | MEDLINE | ID: mdl-27714433

ABSTRACT

The most important goals for the field of immuno-oncology are to improve the response rate and increase the number of tumor indications that respond to immunotherapy, without increasing adverse side effects. One approach to achieve these goals is to use tumor-directed immunotherapy, i.e., to focus the immune activation to the most relevant part of the immune system. This may improve anti-tumor efficacy as well as reduce immune-related adverse events. Tumor-directed immune activation can be achieved by local injections of immune modulators in the tumor area or by directing the immune modulator to the tumor using bispecific antibodies. In this review, we focus on therapies targeting checkpoint inhibitors and co-stimulatory receptors that can generate tumor-specific T cell responses through localized immune activation.


Subject(s)
Antibodies, Bispecific/immunology , Immunologic Factors/immunology , Immunotherapy/methods , Neoplasms/immunology , Neoplasms/therapy , T-Lymphocytes/immunology , Antibodies, Bispecific/pharmacology , Humans , Immunologic Factors/pharmacology
3.
Respir Res ; 13: 53, 2012 Jun 22.
Article in English | MEDLINE | ID: mdl-22726593

ABSTRACT

BACKGROUND: Interactions between Th1 and Th2 immune responses are of importance to the onset and development of allergic disorders. A Toll-like receptor 7 agonist such as AZD8848 may have potential as a treatment for allergic airway disease by skewing the immune system away from a Th2 profile. OBJECTIVE: To evaluate the efficacy and safety of intranasal AZD8848. METHODS: In a placebo-controlled single ascending dose study, AZD8848 (0.3-600 µg) was given intranasally to 48 healthy subjects and 12 patients with allergic rhinitis (NCT00688779). In a placebo-controlled repeat challenge/treatment study, AZD8848 (30 and 60 µg) was given once weekly for five weeks to 74 patients with allergic rhinitis out of season: starting 24 hours after the final dose, daily allergen challenges were given for seven days (NCT00770003). Safety, tolerability, pharmacokinetics, and biomarkers were monitored. During the allergen challenge series, nasal symptoms and lavage fluid levels of tryptase and α2-macroglobulin, reflecting mast cell activity and plasma exudation, were monitored. RESULTS: AZD8848 produced reversible blood lymphocyte reductions and dose-dependent flu-like symptoms: 30-100 µg produced consistent yet tolerable effects. Plasma interleukin-1 receptor antagonist was elevated after administration of AZD8848, reflecting interferon production secondary to TLR7 stimulation. At repeat challenge/treatment, AZD8848 reduced nasal symptoms recorded ten minutes after allergen challenge up to eight days after the final dose. Tryptase and α2-macroglobulin were also reduced by AZD8848. CONCLUSIONS: Repeated intranasal stimulation of Toll-like receptor 7 by AZD8848 was safe and produced a sustained reduction in the responsiveness to allergen in allergic rhinitis. TRIAL REGISTRATION: NCT00688779 and NCT00770003 as indicated above.


Subject(s)
Allergens/administration & dosage , Rhinitis, Allergic, Perennial/prevention & control , Toll-Like Receptor 7/administration & dosage , Administration, Intranasal , Adult , Double-Blind Method , Female , Humans , Male , Rhinitis, Allergic , Toll-Like Receptor 7/agonists , Young Adult
4.
Expert Opin Biol Ther ; 21(12): 1635-1646, 2021 12.
Article in English | MEDLINE | ID: mdl-34043482

ABSTRACT

Introduction: CD40 signaling activates dendritic cells leading to improved T cell priming against tumor antigens. CD40 agonism expands the tumor-specific T cell repertoire and has the potential to increase the fraction of patients that respond to established immunotherapies.Areas covered: This article reviews current as well as emerging CD40 agonist therapies with a focus on antibody-based therapies, including next generation bispecific CD40 agonists. The scientific rationale for different design criteria, binding epitopes, and formats are discussed.Expert opinion: The ability of CD40 agonists to activate dendritic cells and enhance antigen cross-presentation to CD8+ T cells provides an opportunity to elevate response rates of cancer immunotherapies. While there are many challenges left to address, including optimal dose regimen, CD40 agonist profile, combination partners and indications, we are confident that CD40 agonists will play an important role in the challenging task of reprogramming the immune system to fight cancer.


Subject(s)
CD8-Positive T-Lymphocytes , Neoplasms , Antibodies, Monoclonal , CD40 Antigens , Dendritic Cells , Humans , Immunotherapy , Neoplasms/therapy
5.
J Immunother Cancer ; 7(1): 103, 2019 04 11.
Article in English | MEDLINE | ID: mdl-30975201

ABSTRACT

BACKGROUND: The CTLA-4 blocking antibody ipilimumab has demonstrated substantial and durable effects in patients with melanoma. While CTLA-4 therapy, both as monotherapy and in combination with PD-1 targeting therapies, has great potential in many indications, the toxicities of the current treatment regimens may limit their use. Thus, there is a medical need for new CTLA-4 targeting therapies with improved benefit-risk profile. METHODS: ATOR-1015 is a human CTLA-4 x OX40 targeting IgG1 bispecific antibody generated by linking an optimized version of the Ig-like V-type domain of human CD86, a natural CTLA-4 ligand, to an agonistic OX40 antibody. In vitro evaluation of T-cell activation and T regulatory cell (Treg) depletion was performed using purified cells from healthy human donors or cell lines. In vivo anti-tumor responses were studied using human OX40 transgenic (knock-in) mice with established syngeneic tumors. Tumors and spleens from treated mice were analyzed for CD8+ T cell and Treg frequencies, T-cell activation markers and tumor localization using flow cytometry. RESULTS: ATOR-1015 induces T-cell activation and Treg depletion in vitro. Treatment with ATOR-1015 reduces tumor growth and improves survival in several syngeneic tumor models, including bladder, colon and pancreas cancer models. It is further demonstrated that ATOR-1015 induces tumor-specific and long-term immunological memory and enhances the response to PD-1 inhibition. Moreover, ATOR-1015 localizes to the tumor area where it reduces the frequency of Tregs and increases the number and activation of CD8+ T cells. CONCLUSIONS: By targeting CTLA-4 and OX40 simultaneously, ATOR-1015 is directed to the tumor area where it induces enhanced immune activation, and thus has the potential to be a next generation CTLA-4 targeting therapy with improved clinical efficacy and reduced toxicity. ATOR-1015 is also expected to act synergistically with anti-PD-1/PD-L1 therapy. The pre-clinical data support clinical development of ATOR-1015, and a first-in-human trial has started (NCT03782467).


Subject(s)
Antibodies, Bispecific/pharmacology , CTLA-4 Antigen/antagonists & inhibitors , Receptors, OX40/agonists , Urinary Bladder Neoplasms/drug therapy , Animals , Antibodies, Bispecific/therapeutic use , CHO Cells , CTLA-4 Antigen/immunology , Cell Line, Tumor/transplantation , Cricetulus , Disease Models, Animal , Drug Screening Assays, Antitumor , Female , HEK293 Cells , Humans , Male , Mice , Mice, Transgenic , Primary Cell Culture , Proof of Concept Study , Receptors, OX40/genetics , Receptors, OX40/immunology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Urinary Bladder Neoplasms/immunology , Urinary Bladder Neoplasms/pathology
6.
Ther Adv Vaccines Immunother ; 6(1): 3-17, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29998217

ABSTRACT

Following the clinical success of immune checkpoint antibodies targeting CTLA-4, PD-1 or PD-L1 in cancer treatment, bispecific antibodies are now emerging as a growing class of immunotherapies with potential to further improve clinical efficacy and safety. We describe three classes of immunotherapeutic bispecific antibodies: (a) cytotoxic effector cell redirectors; (b) tumor-targeted immunomodulators; and (c) dual immunomodulators. Cytotoxic effector cell redirectors are dominated by T-cell redirecting compounds, bispecific compounds engaging a tumor-associated antigen and the T-cell receptor/CD3 complex, thereby redirecting T-cell cytotoxicity to malignant cells. This is the most established class of bispecific immunotherapies, with two compounds having reached the market and numerous compounds in clinical development. Tumor-targeted immunomodulators are bispecific compounds binding to a tumor-associated antigen and an immunomodulating receptor, such as CD40 or 4-1BB. Such compounds are usually designed to be inactive until binding the tumor antigen, thereby localizing immune stimulation to the tumor environment, while minimizing immune activation elsewhere. This is expected to induce powerful activation of tumor-specific T cells with reduced risk of immune-related adverse events. Finally, dual immunomodulators are bispecific compounds that bind two distinct immunomodulating targets, often combining targeting of PD-1 or PD-L1 with that of LAG-3 or TIM-3. The rationale is to induce superior tumor immunity compared to monospecific antibodies to the same targets. In this review, we describe each of these classes of bispecific antibodies, and present examples of compounds in development.

7.
Regul Pept ; 143(1-3): 118-26, 2007 Oct 04.
Article in English | MEDLINE | ID: mdl-17573135

ABSTRACT

Ghrelin is produced by A-like cells (ghrelin cells) in the mucosa of the acid-producing part of the stomach. The mobilization of ghrelin is stimulated by nutritional deficiency and suppressed by nutritional abundance. In an attempt to identify neurotransmitters and regulatory peptides that may contribute to the physiological, nutrient-related regulation of ghrelin secretion, we challenged the ghrelin cells in situ with a wide variety of candidate messengers, including known neurotransmitters (e.g. acetylcholine, catecholamines), candidate neurotransmitters (e.g. neuropeptides), local tissue hormones (e.g. serotonin, histamine, bradykinin, endothelin), circulating gut hormones (e.g. gastrin, CCK, GIP, neurotensin, PYY, secretin) and other circulating hormones/regulatory peptides (e.g. calcitonin, glucagon, insulin, PTH). Microdialysis probes were placed in the submucosa of the acid-producing part of the rat stomach. Three days later, the putative messenger compounds were administered via the microdialysis probe (reverse microdialysis) at a screening dose of 0.1 mmol l(-1) for regulatory peptides and 0.1 and 1 mmol l(-1) for amines and amino acids. The rats were awake during the experiments. The resulting microdialysate ghrelin concentration was monitored continuously for 3 h (radioimmunoassay), thereby revealing stimulators or inhibitors of ghrelin secretion. Dose-response curves were constructed for each candidate messenger that significantly (p<0.05) affected ghrelin mobilization at the screening dose. Peptides that showed a (non-significant) tendency to affect ghrelin release at the screening dose were also given at a dose of 0.3 or 1 mmol l(-1). Adrenaline, noradrenaline, endothelin and secretin stimulated ghrelin release, while somatostatin and GRP inhibited. Whether these agents act directly or indirectly on the ghrelin cells remains to be investigated. All other candidate messengers were without measurable effects, including acetylcholine, serotonin, histamine, GABA, aspartic acid, glutamic acid, glycine, VIP, PACAP, CGRP, substance P, NPY, PYY, PP, gastrin, CCK, GIP, insulin, glucagon, GLP and glucose.


Subject(s)
Gastric Mucosa/metabolism , Microdialysis/methods , Peptide Hormones/metabolism , Amines/pharmacology , Amino Acids/pharmacology , Animals , Female , Gastric Inhibitory Polypeptide/pharmacology , Gastrins/pharmacology , Gastrointestinal Hormones/pharmacology , Ghrelin , Glucagon/pharmacology , Glucose/pharmacology , Histamine/pharmacology , Insulin/pharmacology , Neuropeptides/pharmacology , Pancreatic Hormones/pharmacology , Peptide YY/pharmacology , Rats , Rats, Sprague-Dawley , Stomach/cytology , Stomach/drug effects
8.
Clin Cancer Res ; 21(5): 1115-26, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25316820

ABSTRACT

PURPOSE: Local administration of immune-activating antibodies may increase the efficacy and reduce the immune-related adverse events associated with systemic immunotherapy of cancer. Here, we report the development and affinity maturation of a fully human agonistic CD40 antibody (IgG1), ADC-1013. EXPERIMENTAL DESIGN: We have used molecular engineering to generate an agonistic antibody with high affinity for CD40. The functional activity of ADC-1013 was investigated in human and murine in vitro models. The in vivo effect was investigated in two separate bladder cancer models, both using human xenograft tumors in immune deficient NSG mice and using a syngeneic bladder cancer model in a novel human CD40 transgenic mouse. RESULTS: Activation of dendritic cells (DC) by ADC-1013 results in upregulation of the costimulatory molecules CD80 and CD86, and secretion of IL12. ADC-1013 also activates DCs from human CD40 transgenic mice, and peptide-pulsed and ADC-1013-stimulated DCs induce antigen-specific T-cell proliferation in vitro. In vivo, treatment with ADC-1013 in a syngeneic bladder cancer model, negative for hCD40, induces significant antitumor effects and long-term tumor-specific immunity. Furthermore, ADC-1013 demonstrates significant antitumor effects in a human bladder cancer transplanted into immunodeficient NSG mice. CONCLUSIONS: Our data demonstrate that ADC-1013 induces long-lasting antitumor responses and immunologic memory mediated by CD40 stimulation. To the best of our knowledge, ADC-1013 represents the first immunomodulatory antibody developed for local immunotherapy of cancer.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , CD40 Antigens/agonists , T-Lymphocytes/immunology , Urinary Bladder Neoplasms/immunology , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal, Humanized , Antibody-Dependent Cell Cytotoxicity/immunology , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , CD40 Antigens/chemistry , CD40 Antigens/genetics , CD40 Antigens/metabolism , Cell Line, Tumor , Dendritic Cells/immunology , Dendritic Cells/metabolism , Disease Models, Animal , Female , Humans , Lymphocyte Activation/immunology , Male , Mice , Mice, Knockout , Models, Molecular , Molecular Conformation , Protein Binding , T-Lymphocytes/metabolism , Tumor Burden/drug effects , Tumor Burden/immunology , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology , Xenograft Model Antitumor Assays
9.
Regul Pept ; 120(1-3): 23-32, 2004 Aug 15.
Article in English | MEDLINE | ID: mdl-15177917

ABSTRACT

Ghrelin, a recently discovered peptide hormone, is produced by endocrine cells in the stomach, the so-called A-like cells. Ghrelin binds to the growth hormone (GH) secretagogue receptor and releases GH. It is claimed to be orexigenic and to control gastric acid secretion and gastric motility. In this study, we examined the effects of ghrelin, des-Gln14-ghrelin, des-octanoyl ghrelin, ghrelin-18, -10 and -5 (and motilin) on gastric emptying in mice and on gastric acid secretion in chronic fistula rats and pylorus-ligated rats. We also examined whether ghrelin affected the activity of the predominant gastric endocrine cell populations, G cells, ECL cells and D cells. Ghrelin and des-Gln14-ghrelin stimulated gastric emptying in a dose-dependent manner while des-octanoyl ghrelin and motilin were without effect. The C-terminally truncated ghrelin fragments were effective but much less potent than ghrelin itself. Ghrelin, des-Gln14-ghrelin and des-octanoyl ghrelin neither stimulated nor inhibited gastric acid secretion, and ghrelin, finally, did not affect secretion from either G cells, ECL cells or D cells.


Subject(s)
Gastric Acid/metabolism , Gastric Emptying/drug effects , Peptide Hormones/pharmacology , Stomach/drug effects , Animals , Dose-Response Relationship, Drug , Female , Gastric Emptying/physiology , Gastric Mucosa/metabolism , Ghrelin , Growth Hormone/pharmacology , Mice , Mice, Inbred Strains , Motilin/pharmacology , Rats , Rats, Sprague-Dawley
10.
Regul Pept ; 106(1-3): 13-8, 2002 Jun 15.
Article in English | MEDLINE | ID: mdl-12047905

ABSTRACT

The existence of an osteotropic hormone (referred to as gastrocalcin) in the ECL cells of the gastric mucosa has been suggested. Both gastrin and an extract of the oxyntic mucosa lower blood Ca(2+) and stimulate Ca(2+) uptake into bone. The ECL cells are known to operate under gastrin control and, conceivably, gastrin lowers blood Ca(2+) indirectly by releasing the hypothetical ECL cell hormone. We have shown earlier that extracts of isolated ECL cells or of the granule/vesicle fraction of the oxyntic mucosa evoke a typical Ca(2+)-mediated second messenger response in osteoblastic cells. In the present study, we characterize this response further. An increase in intracellular inositol 1,4,5-trisphosphate (IP(3)) concentration was observed after treatment of UMR-106.01 osteoblast-like cells with extracts of ECL cells or granule/vesicle-enriched fractions from oxyntic mucosa. Intracellular cyclic adenosine monophosphate (cAMP) concentrations were not affected. Inhibition of phospholipase C (PLC) by U-73122 abolished the increase in [Ca(2+)](i). Preincubation of UMR-106.01 cells with pertussis toxin, which blocks many G-proteins, did not prevent the increases in IP(3) and [Ca(2+)](i). It was also found that the novel peptide hormone ghrelin, produced in the A-like cells of the oxyntic mucosa, did not evoke any Ca(2+) signal in osteoblastic cells. The results indicate that the extracts mediate their effects through a pertussis toxin-insensitive mechanism, and that binding to a receptor leads to activation of PLC and production of IP(3) resulting in increased [Ca(2+)](i). The putative osteotropic hormone is distinct from ghrelin.


Subject(s)
Cell Extracts/pharmacology , Cyclic AMP/metabolism , Enterochromaffin-like Cells/chemistry , Inositol 1,4,5-Trisphosphate/metabolism , Osteoblasts/drug effects , Parietal Cells, Gastric/cytology , Signal Transduction/drug effects , Animals , Calcium/analysis , Calcium/metabolism , Cells, Cultured , Osteoblasts/cytology , Osteoblasts/metabolism , Rats
11.
Regul Pept ; 108(2-3): 73-82, 2002 Oct 15.
Article in English | MEDLINE | ID: mdl-12220729

ABSTRACT

Histamine-producing ECL cells and ghrelin-producing A-like cells are endocrine/paracrine cell populations in the acid-producing part of the rat stomach. While the A-like cells operate independently of gastrin, the ECL cells respond to gastrin with mobilization of histamine and chromogranin A (CGA)-derived peptides, such as pancreastatin. Gastrin is often assumed to be the driving force behind the postnatal development of the gastric mucosa in general and the ECL cells in particular. We tested this assumption by examining the oxyntic mucosa (with ECL cells and A-like cells) in developing rats under the influence of YF476, a cholecystokinin-2 (CCK(2)) receptor antagonist. The drug was administered by weekly subcutaneous injections starting at birth. The body weight gain was not affected. Weaning occurred at days 15-22 in both YF476-treated and age-matched control rats. Circulating gastrin was low at birth and reached adult levels 2 weeks after birth. During and after weaning (but not before), YF476 greatly raised the serum gastrin concentration (because of abolished acid feedback inhibition of gastrin release). The weight of the stomach was unaffected by YF476 during the first 2-3 weeks after birth. From 4 to 5 weeks of age, the weight and thickness of the gastric mucosa were lower in YF476-treated rats than in controls. Pancreastatin-immunoreactive cells (i.e. all endocrine cells in the stomach) and ghrelin-immunoreactive cells (A-like cells) were few at birth and increased gradually in number until 6-8 weeks of age (control rats). At first, YF476 did not affect the development of the pancreastatin-immunoreactive cells, but a few weeks after weaning, the cells were fewer in the YF476 rats. The ECL-cell parameters (oxyntic mucosal histamine and pancreastatin concentrations, the histidine decarboxylase (HDC) activity, the HDC mRNA levels and serum pancreastatin concentration) increased slowly until weaning in both YF476-treated and control rats. From then on, there was a further increase in the ECL-cell parameters in control rats but not in YF476 rats. The postnatal development of the ghrelin cells (i.e. the A-like cells) and of the A-like cell parameters (the oxyntic mucosal ghrelin concentration and the serum ghrelin concentrations) was not affected by YF476 at any point. We conclude that gastrin affects neither the oxyntic mucosa nor the endocrine cells before weaning. After weaning, CCK(2) receptor blockade is associated with a somewhat impaired development of the oxyntic mucosa and the ECL cells. While gastrin stimulation is of crucial importance for the onset of acid secretion during weaning and for the activation of ECL-cell histamine formation and secretion, the mucosal and ECL-cell growth at this stage is only partly gastrin-dependent. In contrast, the development of the A-like cells is independent of gastrin at all stages.


Subject(s)
Aging/physiology , Gastric Mucosa/cytology , Gastric Mucosa/physiology , Gastrins/physiology , Analysis of Variance , Animals , Animals, Newborn , Chromogranin A , Female , Gastric Mucosa/growth & development , Histamine Release , Male , Pancreatic Hormones/analysis , Rats
12.
Regul Pept ; 163(1-3): 24-30, 2010 Aug 09.
Article in English | MEDLINE | ID: mdl-20346991

ABSTRACT

We monitored gastrin release in response to locally applied candidate messengers in intact conscious rats. Earlier studies have been performed on anaesthetized animals, isolated pieces of antrum, or purified preparations of gastrin cells. In this study we created an experimental situation to resemble physiological conditions, using reverse microdialysis to administer regulatory peptides and amines that might affect gastrin secretion. Microdialysis probes were implanted in the submucosa of the antrum of the rat stomach. Three days later, putative messenger compounds were administered via the probe. Their effects on basal (24 h fast) and omeprazole-stimulated (400 micromol/kg/day, 4 days peroral administration) gastrin release were monitored by continuous measurement (3 h) of gastrin in the perfusate (radioimmunoassay). Fasted rats (low microdialysate gastrin, 2.1+/-0.1 pmol l(-1)) were used to study stimulation of gastrin release. Omeprazole-treated rats (high microdialysate gastrin, 95.8+/-6.7 pmol l(-1)) were used to study suppression of gastrin release. The following agents raised the concentration of microdialysate gastrin (peak response): gastrin-releasing peptide (GRP) (11-fold increase at a near-maximal dose), carbachol (5-fold increase), serotonin (2-fold increase) and isoprenaline (20-fold increase). Adrenaline and noradrenaline induced transient but powerful elevation (40- and 20-fold increase). Somatostatin, galanin and bradykinin (at near-maximal doses) suppressed omeprazole-stimulated gastrin release (50% decrease). Calcitonin gene-related peptide, ghrelin, gastric inhibitory peptide, motilin, neurotensin, neuromedin U-25, peptide YY and vasoactive intestinal peptide were without effect on gastrin release, as were aspartate, gamma-aminobutyric acid, glutamate, glycine, dopamine and histamine. The results support the view that G cells operate under neurocrine/paracrine control. They were stimulated by agents present in enteric neurons (GRP, galanin, choline ester and catechol amines) and in gastric endocrine cells (serotonin). They were inhibited by somatostatin (D cell peptide), galanin (neuropeptide) and by the inflammatory agent bradykinin.


Subject(s)
Gastrins/drug effects , Gastrins/metabolism , Microdialysis , Pyloric Antrum/drug effects , Pyloric Antrum/metabolism , Animals , Consciousness , Female , Rats , Rats, Sprague-Dawley
16.
Cell Tissue Res ; 333(3): 405-15, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18592274

ABSTRACT

Histamine in the rat stomach resides in enterochromaffin-like (ECL) cells and mast cells. The ECL cells are peptide-hormone-producing endocrine cells known to release histamine and chromogranin-A-derived peptides (such as pancreastatin) in response to gastrin. Ischemia (induced by clamping of the celiac artery or by gastric submucosal microinfusion of the vasoconstrictor endothelin) mobilizes large amounts of ECL-cell histamine in a burst-like manner. This report examines the ECL-cell response to ischemia and compares it with that induced by gastrin in rats. Arterial clamping (30 min) and gastric submucosal microinfusion (3 h) of endothelin, vasopressin, or adrenaline caused ischemia, manifested as a raised lactate/pyruvate ratio and mucosal damage. Whereas microinfusion of gastrin released both histamine and pancreastatin, ischemia mobilized histamine only. The mucosal concentrations of histamine and pancreastatin, the number and immunostaining intensity of the ECL cells, and the ultrastructure of the ECL cells were unchanged following ischemia. The long-term effects of ischemia and reperfusion (60-90 min) on gastric mucosa were examined in rats treated with the proton pump inhibitor omeprazole for 4 days. The activity of the ECL cells was suppressed (reflected in low histamine-forming capacity) but returned to normal within 1 week, illustrating the ability of the ECL cells to recover. We suggest that ischemia mobilizes cytosolic ECL-cell histamine without affecting the storage of histamine (and pancreastatin) in the secretory organelles and without causing lasting ECL-cell impairment.


Subject(s)
Cell Compartmentation/physiology , Cytosol/metabolism , Enterochromaffin-like Cells/metabolism , Gastric Mucosa/metabolism , Histamine/metabolism , Ischemia/metabolism , Animals , Chromogranin A , Endothelins/pharmacology , Enterochromaffin-like Cells/drug effects , Epinephrine/pharmacology , Female , Gastric Mucosa/drug effects , Gastric Mucosa/pathology , Gastrins/pharmacology , Histamine Release/drug effects , Ischemia/chemically induced , Pancreatic Hormones/metabolism , Rats , Rats, Sprague-Dawley , Secretory Vesicles/metabolism , Vasopressins/pharmacology
17.
Gerodontology ; 24(1): 41-6, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17302930

ABSTRACT

OBJECTIVE: The aim of this study was to re-evaluate data about oral status, mastication and nutrition in elderly men in Malmö, Sweden, recorded in 1985-1987, to assess associations between inadequate dietary habits, oral conditions and masticatory function. MATERIALS AND METHODS: Four hundred and eighty-one men, aged 67-68, participated in a comprehensive health examination, including tooth and denture status and masticatory tests. A separate study of dietary habits and nutritional status was made. Ninety-five men had inadequate dietary habits. The databases of dental/denture status, mastication, nutritional status and social network factors were re-evaluated for assessment of associations. RESULTS: No significant differences between those with adequate or inadequate nutrition were found with regard to the number of teeth, occlusal contacts or removable dentures. Also self-assessed chewing did not show any differences. CONCLUSION: Inadequate dietary habits were independent of teeth and denture status. Some correlations to social network conditions could be identified. Overweight, obesity, low physical activity and high alcohol intake were more common among those with inadequate nutritional intake.


Subject(s)
Feeding Behavior , Mastication/physiology , Aged , Alcohol Drinking , Cohort Studies , Dental Occlusion , Dentition , Denture, Complete , Denture, Partial , Humans , Male , Motor Activity/physiology , Nutritional Physiological Phenomena , Nutritional Status , Obesity/classification , Oral Health , Overweight/physiology , Social Support , Sweden
18.
Chem Pharm Bull (Tokyo) ; 55(4): 635-7, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17409561

ABSTRACT

In our continuing research on cytotoxic components from the Formosan pteridophyte Thelypteris torresiana (GAUD.) ALSTONONE, two new compounds, a novel flavonoid, flavotorresin (1), and a flavonoid diglycoside, multiflorin C (2), along with five known compounds, were isolated. The structural elucidation was established on the basis of spectroscopic data analysis. The possible biosynthetic pathway of the flavonoids from this fern is summarized.


Subject(s)
Ferns/chemistry , Flavonoids/isolation & purification , Flavonoids/chemistry , Spectrum Analysis/methods
19.
Hum Mol Genet ; 15(10): 1713-21, 2006 May 15.
Article in English | MEDLINE | ID: mdl-16613897

ABSTRACT

Huntington's disease (HD) is characterized by a triad of motor, psychiatric and cognitive symptoms. Although many of these symptoms are likely to be related to central nervous system pathology, others may be due to changes in peripheral tissues. The R6/2 mouse, a transgenic model of HD expressing exon 1 of the human HD gene, develops progressive alterations in the hypothalamic-pituitary-adrenal axis, reminiscent of a Cushing-like syndrome. We observed muscular atrophy, reduced bone mineral density, abdominal fat accumulation and insulin resistance in the mice. All these changes could be consequences of increased glucocorticoid levels. Indeed, hypertrophy of the adrenal cortex and a progressive increase in serum and urine corticosterone levels were found in R6/2 mice. In addition, the intermediate pituitary lobe was markedly enlarged and circulating adreno-corticotrophic hormone (ACTH) increased. Under normal conditions dopamine represses the ACTH expression. In the R6/2 mice, however, the expression of pituitary dopamine D2 receptors was reduced by half, possibly explaining the increase in ACTH. Urinary samples from 82 HD patients and 68 control subjects were analysed for cortisol: in accord with the observations in the R6/2 mice, urinary cortisol increased in parallel with disease progression. This progressive increase in cortisol may contribute to the clinical symptoms, such as muscular wasting, mood changes and some of the cognitive deficits that occur in HD.


Subject(s)
Corticosterone/blood , Huntington Disease/pathology , Hypothalamo-Hypophyseal System/pathology , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Pituitary-Adrenal System/pathology , Adrenocorticotropic Hormone/blood , Adrenocorticotropic Hormone/urine , Adult , Animals , Body Fat Distribution , Bone Density , Corticosterone/urine , Disease Models, Animal , Dopamine/physiology , Female , Humans , Huntingtin Protein , Huntington Disease/metabolism , Hydrocortisone/blood , Hydrocortisone/urine , Hypothalamo-Hypophyseal System/physiopathology , Insulin Resistance , Male , Mice , Mice, Transgenic , Middle Aged , Muscular Atrophy/metabolism , Muscular Atrophy/pathology , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Pituitary-Adrenal System/physiopathology , Receptors, Dopamine D2/metabolism
20.
Am J Physiol Gastrointest Liver Physiol ; 288(5): G1084-90, 2005 May.
Article in English | MEDLINE | ID: mdl-15662050

ABSTRACT

Microdialysis was used to study how ischemia-evoked gastric mucosal injury affects rat stomach histamine, which resides in enterochromaffin-like (ECL) cells and mast cells. A microdialysis probe was inserted into the gastric submucosa, and the celiac artery was clamped (30 min), followed by removal of the clamp. Microdialysate histamine was determined by enzyme-linked immunosorbent assay. In addition, we studied the long-term effects of ischemia on the oxyntic mucosal histidine decarboxylase activity in omeprazole-treated rats. Gastric mucosal lesions induced by the ischemia were enlarged on removal of the clamp. The microdialysate histamine concentration increased immediately on clamping (50-fold rise within 30 min) and declined promptly after the clamp was removed. In contrast, histidine decarboxylase activity of the ECL cells was lowered by the ischemia and returned to preischemic values 9 days later. Mast cell-deficient rats responded to ischemia-reperfusion much like wild-type rats with respect to histamine mobilization. Pretreatment with the irreversible inhibitor of histidine decarboxylase, alpha-fluoromethylhistidine, which is known to eliminate histamine from ECL cells, prevented the rise in microdialysate histamine. Pharmacological blockade of acid secretion (cimetidine or omeprazole) prevented the lesions induced by ischemia-reperfusion insult but not the mobilization of histamine. In conclusion, ischemia of the celiac artery mobilizes large amounts of histamine from ECL cells, which occurs independently of the gross mucosal lesions. The prompt reduction of the mucosal histidine decarboxylase activity in response to ischemia probably reflects ECL cell damage. The lesions develop not because of mobilization of histamine per se but because of ischemia plus reperfusion plus gastric acid.


Subject(s)
Enterochromaffin-like Cells/metabolism , Gastric Mucosa/blood supply , Gastric Mucosa/metabolism , Histamine/metabolism , Ischemia/metabolism , Animals , Cimetidine/pharmacology , Enterochromaffin-like Cells/drug effects , Gastric Mucosa/cytology , Histamine H2 Antagonists/pharmacology , Male , Mast Cells/physiology , Methylhistidines/pharmacology , Omeprazole/pharmacology , Proton Pump Inhibitors , Rats , Rats, Wistar , Reperfusion Injury/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL