Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Zookeys ; 1193: 95-110, 2024.
Article in English | MEDLINE | ID: mdl-38463757

ABSTRACT

New data are presented for the potter wasp genus Orancistrocerus van der Vecht (Eumeninae, Odynerini) occurring in Vietnam. Two species are described as new to science: Orancistrocerusthanhnhatsp. nov. and O.thanghensp. nov.Orancistrocerusaterrimuserythropus van der Vecht is synonymized with Orancistrocerusaterrimusaterrimus (de Saussure); the male genitalia of this species are described for the first time. An updated key is presented to all species of the genus.

2.
Insects ; 14(6)2023 Jun 06.
Article in English | MEDLINE | ID: mdl-37367346

ABSTRACT

Cassava is a valuable export commodity crop that is often attacked by pests, causing economic losses for this crop. The papaya mealybug Paracoccus marginatus has become a major pest of cassava in Vietnam. The parasitoid wasp Acerophagus papayae has been demonstrated to be the most efficient parasitoid wasp for controlling P. marginatus in many regions. We observed the occurrence of A. papayae in Vietnam, studied the biological characteristics of A. papayae, and investigated its parasitic activity on P. marginatus. The results showed that A. papayae occurred more frequently than Anagyrus loecki, another known parasitoid of P. marginatus. The lifespan of A. papayae was approximately 16 days. In the absence of hosts, a 50% honey solution was an essential diet to increase the longevity of both female and male of A. papayae. The second instar of P. marginatus was a suitable host stage for parasitism by A. papayae. Female A. papayae laid approximately 60.8 eggs within 17 days, mostly during the first 6 to 7 days. These findings suggest that A. papayae has the potential to control P. marginatus, and could inform the development of more effective pest management strategies for cassava crops in Vietnam and other regions affected by this pest.

3.
Sci Rep ; 13(1): 6365, 2023 04 19.
Article in English | MEDLINE | ID: mdl-37076660

ABSTRACT

The use of gnobiotic brine shrimp (Artemia spp.) for ecotoxicology and bacteria-host interaction studies is common. However, requirements for axenic culture and matrix effects of seawater media can be an obstacle. Thus, we investigated the hatching ability of Artemia cysts on a novel sterile Tryptic Soy Agar (TSA) medium. Herein, we demonstrate for the first time that Artemia cysts can hatch on a solid medium without liquid, which offers practical advantages. We further optimized the culture conditions for temperature and salinity and assessed this culture system for toxicity screening of silver nanoparticles (AgNPs) across multiple biological endpoints. Results revealed that maxima hatching (90%) of embryos occurred at 28 °C and without addition of sodium chloride. When capsulated cysts were cultured on TSA solid medium Artemia were negatively impacted by AgNPs at 30-50 mgL-1 in terms of the embryo hatching ratio (47-51%), umbrella- to nauplii-stage transformation ratio (54-57%), and a reduction in nauplii-stage growth (60-85% of normal body length). At 50-100 mgL-1 AgNPs and higher, evidence of damage to lysosomal storage was recorded. At 500 mgL-1 AgNPs, development of the eye was inhibited and locomotory behavior impeded. Our study reveals that this new hatching method has applications in ecotoxicology studies and provides an efficient means to control axenic requirements to produce gnotobiotic brine shrimp.


Subject(s)
Metal Nanoparticles , Animals , Metal Nanoparticles/toxicity , Silver/toxicity , Artemia , Agar/pharmacology , Ecotoxicology , Culture Media/pharmacology
4.
Sci Data ; 9(1): 432, 2022 07 21.
Article in English | MEDLINE | ID: mdl-35864125

ABSTRACT

One of the effects of COVID-19 pandemic is a rapidly growing and changing stream of publications to inform clinicians, researchers, policy makers, and patients about the health, socio-economic, and cultural consequences of the pandemic. Managing this information stream manually is not feasible. Automatic Question Answering can quickly bring the most salient points to the user's attention. Leveraging a collection of scientific articles, government websites, relevant news articles, curated social media posts, and questions asked by researchers, clinicians, and the general public, we developed a dataset to explore automatic Question Answering for multiple stakeholders. Analysis of questions asked by various stakeholders shows that while information needs of experts and the public may overlap, satisfactory answers to these questions often originate from different information sources or benefit from different approaches to answer generation. We believe that this dataset has the potential to support the development of question answering systems not only for epidemic questions, but for other domains with varying expertise such as legal or finance.


Subject(s)
COVID-19 , Pandemics , Humans
5.
Int J Biol Macromol ; 209(Pt A): 441-451, 2022 Jun 01.
Article in English | MEDLINE | ID: mdl-35413313

ABSTRACT

The study aims to (a) enhance the solubility of a poorly soluble drug by optimization of nanocrystal formulation using the top-down approach and (b) modify the release profile of this drug, which exhibits a short elimination half-life, by the integration of a fast-release phase containing the optimized nanocrystals and a sustained-release phase in a compression-coated tablet. Nanocrystals of the model drug (lornoxicam; LNX) was prepared by simultaneous application of jet-milling and ball-milling techniques. Investigation of the precipitation inhibition capacity, thermal property, and interaction of different polymers with the drug revealed polyvinyl pyrrolidone K30 (PVP) as the most effective stabilizer for nanocrystals. The immediate-release layer containing the optimized nanocrystals (size of 279.5 ± 11.25 nm and polydispersity index of 0.204 ± 0.01) was then compressed on a zero-order sustained-release matrix core using different derivatives of hydroxypropyl methylcellulose (HPMC). Application of the Design of Experiment approach (DoE) was applied to optimize the formulation of tablet. Analysis of drug concentration in dog plasma by liquid chromatography-tandem mass spectrometry demonstrated an improvement in the release behavior of LNX from the optimal compression-coated tablet integrating a HPMC-based sustained release matrix core and a PVP-stabilized lornoxicam nanocrystals coating layer compared to the reference product.


Subject(s)
Methylcellulose , Nanoparticles , Animals , Delayed-Action Preparations/chemistry , Dogs , Hypromellose Derivatives , Methylcellulose/chemistry , Piroxicam/analogs & derivatives , Povidone , Solubility , Tablets
6.
Immunohorizons ; 3(10): 498-510, 2019 10 21.
Article in English | MEDLINE | ID: mdl-31636084

ABSTRACT

Immune cells express γ-aminobutyric acid receptors (GABA-R), and GABA administration can inhibit effector T cell responses in models of autoimmune disease. The pharmacokinetic properties of GABA, however, may be suboptimal for clinical applications. The amino acid homotaurine is a type A GABA-R (GABAA-R) agonist with good pharmacokinetics and appears safe for human consumption. In this study, we show that homotaurine inhibits in vitro T cell proliferation to a similar degree as GABA but at lower concentrations. In vivo, oral homotaurine treatment had a modest ability to reverse hyperglycemia in newly hyperglycemic NOD mice but was ineffective after the onset of severe hyperglycemia. In severely diabetic NOD mice, the combination of homotaurine and low-dose anti-CD3 treatment significantly increased 1) disease remission, 2) the percentages of splenic CD4+and CD8+ regulatory T cells compared with anti-CD3 alone, and 3) the frequencies of CD4+ and CD8+ regulatory T cells in the pancreatic lymph nodes compared with homotaurine monotherapy. Histological examination of their pancreata provided no evidence of the large-scale GABAA-R agonist-mediated replenishment of islet ß-cells that has been reported by others. However, we did observe a few functional islets in mice that received combined therapy. Thus, GABAA-R activation enhanced CD4+and CD8+ regulatory T cell responses following the depletion of effector T cells, which was associated with the preservation of some functional islets. Finally, we observed that homotaurine treatment enhanced ß-cell replication and survival in a human islet xenograft model. Hence, GABAA-R agonists, such as homotaurine, are attractive candidates for testing in combination with other therapeutic agents in type 1 diabetes clinical trials.


Subject(s)
CD4-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/drug effects , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 1/drug therapy , GABA Agonists/pharmacology , GABA Agonists/therapeutic use , Muromonab-CD3/therapeutic use , Taurine/analogs & derivatives , Animals , Blood Glucose/drug effects , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Type 1/immunology , Drug Synergism , Female , Humans , Insulin-Secreting Cells/drug effects , Islets of Langerhans Transplantation , Mice, Inbred NOD , Mice, SCID , Muromonab-CD3/pharmacology , Taurine/pharmacology , Taurine/therapeutic use
7.
J Diabetes Res ; 2019: 5783545, 2019.
Article in English | MEDLINE | ID: mdl-30937314

ABSTRACT

A major goal of T1D research is to develop new approaches to increase ß-cell mass and control autoreactive T cell responses. GABAA-receptors (GABAA-Rs) are promising drug targets in both those regards due to their abilities to promote ß-cell replication and survival, as well as inhibit autoreactive T cell responses. We previously showed that positive allosteric modulators (PAMs) of GABAA-Rs could promote rat ß-cell line INS-1 and human islet cell replication in vitro. Here, we assessed whether treatment with alprazolam, a widely prescribed GABAA-R PAM, could promote ß-cell survival and replication in human islets after implantation into NOD/scid mice. We observed that alprazolam treatment significantly reduced human islet cell apoptosis following transplantation and increased ß-cell replication in the xenografts. Evidently, the GABAA-R PAM works in conjunction with GABA secreted from ß-cells to increase ß-cell survival and replication. Treatment with both the PAM and GABA further enhanced human ß-cell replication. Alprazolam also augmented the ability of suboptimal doses of GABA to inhibit antigen-specific T cell responses in vitro. Thus, combined GABAA-R agonist and PAM treatment may help control inflammatory immune responses using reduced drug dosages. Together, these findings suggest that GABAA-R PAMs represent a promising drug class for safely modulating islet cells toward beneficial outcomes to help prevent or reverse T1D and, together with a GABAA-R agonist, may have broader applications for ameliorating other disorders in which inflammation contributes to the disease process.


Subject(s)
Alprazolam/pharmacology , Cell Survival , Inflammation , Insulin-Secreting Cells/cytology , Receptors, GABA/metabolism , T-Lymphocytes/metabolism , Animals , Apoptosis , Cell Division , Cell Proliferation , Humans , Islets of Langerhans , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Muramidase , gamma-Aminobutyric Acid/pharmacology
8.
Sci Rep ; 9(1): 4269, 2019 03 12.
Article in English | MEDLINE | ID: mdl-30862859

ABSTRACT

There has been considerable debate as to whether obesity can act as an accelerator of type 1 diabetes (T1D). We assessed this possibility using transgenic mice (MIP-TF mice) whose ß-cells express enhanced green fluorescent protein (EGFP). Infecting these mice with EGFP-expressing murine herpes virus-68 (MHV68-EGFP) caused occasional transient elevation in their blood glucose, peri-insulitis, and Th1 responses to EGFP which did not spread to other ß-cell antigens. We hypothesized that obesity-related systemic inflammation and ß-cell stress could exacerbate the MHV68-EGFP-induced ß-cell autoreactivity. We crossed MIP-TF mice with Avy mice which develop obesity and provide models of metabolic disease alongside early stage T2D. Unlike their MIP-TF littermates, MHV68-EGFP-infected Avy/MIP-TF mice developed moderate intra-insulitis and transient hyperglycemia. MHV68-EGFP infection induced a more pronounced intra-insulitis in older, more obese, Avy/MIP-TF mice. Moreover, in MHV68-EGFP-infected Avy/MIP-TF mice, Th1 reactivity spread from EGFP to other ß-cell antigens. Thus, the spreading of autoreactivity among ß-cell antigens corresponded with the transition from peri-insulitis to intra-insulitis and occurred in obese Avy/MIP-TF mice but not lean MIP-TF mice. These observations are consistent with the notion that obesity-associated systemic inflammation and ß-cell stress lowers the threshold necessary for T cell autoreactivity to spread from EGFP to other ß-cell autoantigens.


Subject(s)
Agouti Signaling Protein/genetics , Diabetes Mellitus, Type 1/immunology , Insulin-Secreting Cells/immunology , Obesity/immunology , T-Lymphocytes/immunology , Animals , Autoantigens/immunology , Autoantigens/metabolism , Autoimmunity/genetics , Blood Glucose/analysis , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/diagnosis , Diabetes Mellitus, Type 1/genetics , Diet, High-Fat/adverse effects , Disease Models, Animal , Humans , Insulin-Secreting Cells/metabolism , Mice , Mice, Transgenic , Mutation , Obesity/complications , Obesity/genetics , Obesity/pathology
9.
Drug Saf ; 42(5): 671-682, 2019 05.
Article in English | MEDLINE | ID: mdl-30478823

ABSTRACT

INTRODUCTION: Despite the numerous studies investigating drug-induced anaphylaxis (DIA), understanding and quantitative data analysis in developing countries remain limited. The aim of our study is to describe and quantify DIA using the National Pharmacovigilance Database of Vietnam (NPDV). METHODS: Spontaneous reporting of adverse drug reactions (ADRs) recorded between 2010 and 2016 were retrospectively analysed to identify DIA reports. The trend and characteristics of DIA cases were described. Multivariate disproportionality analysis was used for signal generation. RESULTS: Overall, 4873 DIA cases (13.2% of total ADRs) were recorded in the NPDV, 111 of which resulted in death (82% of total ADR-induced deaths) over a 7-year period. There was a remarkable increase in DIA reporting over time (p < 0.001). The incidence rates of DIA reporting per total ADRs and per 100,000 inhabitants remained high (mean rates [95% CI] of 12.06 [9.88-14.24] and 0.77 [0.33-1.20], respectively). Concerning suspected drugs, systemic antibiotics (n = 3318, 68%) were mostly reported with a reporting odds ratio (ROR) and 95% CI of 2.35 [2.20-2.51]. In the case of antibiotic-induced anaphylaxis, the third-generation cephalosporins were predominant (n = 1961, 40.2%, ROR 2.39 [2.24-2.55]). We also noted drugs generally associated with DIA such as contrast agents (ROR 2.43 [2.04-2.88]) and anaesthetics (ROR 4.02 [3.30-4.89]). Furthermore, unexpected signals were observed for alpha-chymotrypsin (ROR 1.75 [1.23-2.44]) and amoxicillin/sulbactam (ROR 1.59 [1.18-2.10]), uncommonly reported in western countries. CONCLUSION: In recent years, cases of drug-induced DIA have increased in Vietnam, mostly due to antibiotics and third-generation cephalosporins. The inappropriate use of these drugs should be taken into account. Our findings also highlighted typical Vietnamese signals for alpha-chymotrypsin- and amoxicillin/sulbactam-induced anaphylaxis, which may relate to a specific sociological context in resource-limited countries.


Subject(s)
Adverse Drug Reaction Reporting Systems , Anaphylaxis/epidemiology , Databases, Factual , Pharmacovigilance , Humans , Vietnam/epidemiology
10.
Sci Rep ; 8(1): 16555, 2018 11 08.
Article in English | MEDLINE | ID: mdl-30410049

ABSTRACT

There is a need for treatments that can safely promote regulatory lymphocyte responses. T cells express GABA receptors (GABAA-Rs) and GABA administration can inhibit Th1-mediated processes such as type 1 diabetes and rheumatoid arthritis in mouse models. Whether GABAA-R agonists can also inhibit Th17-driven processes such as experimental autoimmune encephalomyelitis (EAE), a model of multiple sclerosis (MS), is an open question. GABA does not pass through the blood-brain barrier (BBB) making it ill-suited to inhibit the spreading of autoreactivity within the CNS. Homotaurine is a BBB-permeable amino acid that antagonizes amyloid fibril formation and was found to be safe but ineffective in long-term Alzheimer's disease clinical trials. Homotaurine also acts as GABAA-R agonist with better pharmacokinetics than that of GABA. Working with both monophasic and relapsing-remitting mouse models of EAE, we show that oral administration of homotaurine can (1) enhance CD8+CD122+PD-1+ and CD4+Foxp3+ Treg, but not Breg, responses, (2) inhibit autoreactive Th17 and Th1 responses, and (3) effectively ameliorate ongoing disease. These observations demonstrate the potential of BBB-permeable GABAA-R agonists as a new class of treatment to enhance CD8+ and CD4+ Treg responses and limit Th17 and Th1-medaited inflammation in the CNS.


Subject(s)
GABA Agonists/administration & dosage , Multiple Sclerosis/drug therapy , Taurine/analogs & derivatives , Administration, Oral , Animals , Blood-Brain Barrier/metabolism , Disease Models, Animal , GABA Agonists/pharmacology , Mice , Mice, Inbred C57BL , Multiple Sclerosis/immunology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Taurine/administration & dosage , Taurine/pharmacology , Th1 Cells/drug effects , Th1 Cells/immunology , Th17 Cells/drug effects , Th17 Cells/immunology
11.
J Diabetes Res ; 2017: 6403539, 2017.
Article in English | MEDLINE | ID: mdl-29018828

ABSTRACT

The activation of ß-cell's A- and B-type gamma-aminobutyric acid receptors (GABAA-Rs and GABAB-Rs) can promote their survival and replication, and the activation of α-cell GABAA-Rs promotes their conversion into ß-cells. However, GABA and the most clinically applicable GABA-R ligands may be suboptimal for the long-term treatment of diabetes due to their pharmacological properties or potential side-effects on the central nervous system (CNS). Lesogaberan (AZD3355) is a peripherally restricted high-affinity GABAB-R-specific agonist, originally developed for the treatment of gastroesophageal reflux disease (GERD) that appears to be safe for human use. This study tested the hypothesis that lesogaberan could be repurposed to promote human islet cell survival and ß-cell replication. Treatment with lesogaberan significantly enhanced replication of human islet cells in vitro, which was abrogated by a GABAB-R antagonist. Immunohistochemical analysis of human islets that were grafted into immune-deficient mice revealed that oral treatment with lesogaberan promoted human ß-cell replication and islet cell survival in vivo as effectively as GABA (which activates both GABAA-Rs and GABAB-Rs), perhaps because of its more favorable pharmacokinetics. Lesogaberan may be a promising drug candidate for clinical studies of diabetes intervention and islet transplantation.


Subject(s)
Apoptosis/drug effects , Drug Repositioning , GABA-B Receptor Agonists/pharmacology , Hypoglycemic Agents/pharmacology , Insulin-Secreting Cells/drug effects , Islets of Langerhans/drug effects , Phosphinic Acids/pharmacology , Propylamines/pharmacology , Animals , Cell Proliferation/drug effects , Cell Survival/drug effects , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/surgery , GABA-B Receptor Agonists/chemistry , GABA-B Receptor Agonists/therapeutic use , GABA-B Receptor Antagonists/pharmacology , Humans , Hypoglycemic Agents/antagonists & inhibitors , Hypoglycemic Agents/therapeutic use , Immunohistochemistry , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Islets of Langerhans Transplantation , Mice, SCID , Phosphinic Acids/antagonists & inhibitors , Phosphinic Acids/therapeutic use , Propylamines/antagonists & inhibitors , Propylamines/therapeutic use , Random Allocation , Tissue Banks , Tissue Culture Techniques , Transplantation, Heterotopic
12.
Sci Rep ; 7(1): 374, 2017 03 23.
Article in English | MEDLINE | ID: mdl-28336918

ABSTRACT

A key goal of diabetes research is to develop treatments to safely promote human ß-cell replication. It has recently become appreciated that activation of γ-aminobutyric acid receptors (GABA-Rs) on ß-cells can promote their survival and replication. A number of positive allosteric modulators (PAMs) that enhance GABA's actions on neuronal GABAA-Rs are in clinical use. Repurposing these GABAA-R PAMs to help treat diabetes is theoretically appealing because of their safety and potential to enhance the ability of GABA, secreted from ß-cells, or exogenously administered, to promote ß-cell replication and survival. Here, we show that clinically applicable GABAA-R PAMs can increase significantly INS-1 ß-cell replication, which is enhanced by exogenous GABA application. Furthermore, a GABAA-R PAM promoted human islet cell replication in vitro. This effect was abrogated by a GABAA-R antagonist. The combination of a PAM and low levels of exogenous GABA further increased human islet cell replication. These findings suggest that PAMs may potentiate the actions of GABA secreted by islet ß-cells on GABAA-Rs and provide a new class of drugs for diabetes treatment. Finally, our findings may explain a past clinical observation of a GABAA-R PAM reducing HbA1c levels in diabetic patients.


Subject(s)
Benzodiazepines/pharmacology , Cell Division/drug effects , Cell Proliferation/drug effects , Insulin-Secreting Cells/drug effects , Receptors, GABA-A/physiology , gamma-Aminobutyric Acid/biosynthesis , Allosteric Regulation , Alprazolam/pharmacology , Animals , Carrier Proteins/metabolism , Clonazepam/pharmacology , GABA Modulators/pharmacology , GABA-A Receptor Antagonists/administration & dosage , Humans , Insulin-Secreting Cells/physiology , Islets of Langerhans/drug effects , Islets of Langerhans/physiology , Mice , Midazolam/pharmacology , Rats , Receptors, GABA-A/metabolism , gamma-Aminobutyric Acid/administration & dosage
13.
Diabetes ; 63(9): 3128-34, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25146474

ABSTRACT

Antigen-based therapies (ABTs) fail to restore normoglycemia in newly diabetic NOD mice, perhaps because too few ß-cells remain by the time that ABT-induced regulatory responses arise and spread. We hypothesized that combining a fast-acting anti-inflammatory agent with an ABT could limit pathogenic responses while ABT-induced regulatory responses arose and spread. γ-Aminobutyric acid (GABA) administration can inhibit inflammation, enhance regulatory T-cell (Treg) responses, and promote ß-cell replication in mice. We examined the effect of combining a prototypic ABT, proinsulin/alum, with GABA treatment in newly diabetic NOD mice. Proinsulin/alum monotherapy failed to correct hyperglycemia, while GABA monotherapy restored normoglycemia for a short period. Combined treatment restored normoglycemia in the long term with apparent permanent remission in some mice. Proinsulin/alum monotherapy induced interleukin (IL)-4- and IL-10-secreting T-cell responses that spread to other ß-cell autoantigens. GABA monotherapy induced moderate IL-10 (but not IL-4) responses to ß-cell autoantigens. Combined treatment synergistically reduced spontaneous type 1 T-helper cell responses to autoantigens, ABT-induced IL-4 and humoral responses, and insulitis, but enhanced IL-10 and Treg responses and promoted ß-cell replication in the islets. Thus, combining ABT with GABA can inhibit pathogenic T-cell responses, induce Treg responses, promote ß-cell replication, and effectively restore normoglycemia in newly diabetic NOD mice. Since these treatments appear safe for humans, they hold promise for type 1 diabetes intervention.


Subject(s)
Alum Compounds/administration & dosage , Autoimmunity/drug effects , Diabetes Mellitus, Type 1/drug therapy , Proinsulin/administration & dosage , T-Lymphocytes, Regulatory/immunology , Animals , Autoimmunity/immunology , Blood Glucose/metabolism , Cell Division/drug effects , Diabetes Mellitus, Type 1/immunology , Drug Therapy, Combination , Female , Interleukin-10/biosynthesis , Interleukin-4/biosynthesis , Mice , Mice, Inbred NOD
14.
Environ Manage ; 54(2): 331-45, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24894008

ABSTRACT

Many countries are confronting climate change that threatens agricultural production and farmers' lives. Farmers' perceived risks of climate change and factors influencing those perceived risks are critical to their adaptive behavior and well-planned adaptation strategies. However, there is limited understanding of these issues. In this paper, we attempt to quantitatively measure farmers' perceived risks of climate change and explore the influences of risk experience, information, belief in climate change, and trust in public adaptation to those perceived risks. Data are from structured interviews with 598 farmers in the Mekong Delta. The study shows that perceived risks to production, physical health, and income dimensions receive greater priority while farmers pay less attention to risks to happiness and social relationships. Experiences of the events that can be attributed to climate change increase farmers' perceived risks. Information variables can increase or decrease perceived risks, depending on the sources of information. Farmers who believe that climate change is actually happening and influencing their family's lives, perceive higher risks in most dimensions. Farmers who think that climate change is not their concern but the government's, perceive lower risks to physical health, finance, and production. As to trust in public adaptation, farmers who believe that public adaptive measures are well co-ordinated, perceive lower risks to production and psychology. Interestingly, those who believe that the disaster warning system is working well, perceive higher risks to finance, production, and social relationships. Further attention is suggested for the quality, timing, and channels of information about climate change and adaptation.


Subject(s)
Agriculture/methods , Climate Change , Public Opinion , Agriculture/statistics & numerical data , Health Status , Humans , Income , Interviews as Topic , Regression Analysis , Risk Assessment/methods , Risk Assessment/trends , Vietnam
15.
Diabetes ; 62(11): 3760-5, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23995958

ABSTRACT

γ-Aminobutyric acid (GABA) has been shown to inhibit apoptosis of rodent ß-cells in vitro. In this study, we show that activation of GABAA receptors (GABAA-Rs) or GABAB-Rs significantly inhibits oxidative stress-related ß-cell apoptosis and preserves pancreatic ß-cells in streptozotocin-rendered hyperglycemic mice. Moreover, treatment with GABA, or a GABAA-R- or GABAB-R-specific agonist, inhibited human ß-cell apoptosis following islet transplantation into NOD/scid mice. Accordingly, activation of GABAA-Rs and/or GABAB-Rs may be a useful adjunct therapy for human islet transplantation. GABA-R agonists also promoted ß-cell replication in hyperglycemic mice. While a number of agents can promote rodent ß-cell replication, most fail to provide similar activities with human ß-cells. In this study, we show that GABA administration promotes ß-cell replication and functional recovery in human islets following implantation into NOD/scid mice. Human ß-cell replication was induced by both GABAA-R and GABAB-R activation. Hence, GABA regulates both the survival and replication of human ß-cells. These actions, together with the anti-inflammatory properties of GABA, suggest that modulation of peripheral GABA-Rs may represent a promising new therapeutic strategy for improving ß-cell survival following human islet transplantation and increasing ß-cells in patients with diabetes.


Subject(s)
Apoptosis/drug effects , Insulin-Secreting Cells/drug effects , gamma-Aminobutyric Acid/pharmacology , Animals , Baclofen/pharmacology , Cell Survival/drug effects , Diabetes Mellitus, Experimental , GABA-A Receptor Agonists/pharmacology , Heterografts , Humans , Insulin-Secreting Cells/pathology , Islets of Langerhans Transplantation , Male , Mice , Mice, Inbred NOD , Muscimol/pharmacology , Receptors, GABA-A/physiology
16.
J Neurosci Res ; 91(10): 1292-302, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23907992

ABSTRACT

We previously showed that, in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of Parkinson's disease (PD), vaccination with bacillus Calmette-Guerin (BCG) prior to MPTP exposure limited the loss of striatal dopamine (DA) and dopamine transporter (DAT) and prevented the activation of nigral microglia. Here, we conducted BCG dose studies and investigated the mechanisms underlying BCG vaccination's neuroprotective effects in this model. We found that a dose of 1 × 10(6) cfu BCG led to higher levels of striatal DA and DAT ligand binding (28% and 42%, respectively) in BCG-vaccinated vs. unvaccinated MPTP-treated mice, but without a significant increase in substantia nigra tyrosine hydroxylase-staining neurons. Previous studies showed that BCG can induce regulatory T cells (Tregs) and that Tregs are neuroprotective in models of neurodegenerative diseases. However, MPTP is lymphotoxic, so it was unclear whether Tregs were maintained after MPTP treatment and whether a relationship existed between Tregs and the preservation of striatal DA system integrity. We found that, 21 days post-MPTP treatment, Treg levels in mice that had received BCG prior to MPTP were threefold greater than those in MPTP-only-treated mice and elevated above those in saline-only-treated mice, suggesting that the persistent BCG infection continually promoted Treg responses. Notably, the magnitude of the Treg response correlated positively with both striatal DA levels and DAT ligand binding. Therefore, BCG vaccine-mediated neuroprotection is associated with Treg levels in this mouse model. Our results suggest that BCG-induced Tregs could provide a new adjunctive therapeutic approach to ameliorating pathology associated with PD and other neurodegenerative diseases.


Subject(s)
BCG Vaccine/immunology , Neuroprotective Agents/immunology , Parkinsonian Disorders/immunology , Parkinsonian Disorders/prevention & control , T-Lymphocytes, Regulatory/immunology , Animals , BCG Vaccine/administration & dosage , Brain/immunology , Brain/pathology , Chromatography, High Pressure Liquid , Disease Models, Animal , Flow Cytometry , Male , Mice , Mice, Inbred C57BL , Neuroprotective Agents/administration & dosage
17.
J Neuroimmunol ; 247(1-2): 1-8, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22503373

ABSTRACT

We studied cultured hippocampal neurons from embryonic wildtype, major histocompatibility complex class I (MHCI) heavy chain-deficient (K(b)D(b)-/-) and NSE-D(b) (which have elevated neuronal MHCI expression) C57BL/6 mice. K(b)D(b)-/- neurons displayed slower neuritogenesis and establishment of polarity, while NSE-D(b) neurons had faster neurite outgrowth, more primary neurites, and tended to have accelerated polarization. Additional studies with ß2M-/- neurons, exogenous ß2M, and a self-MHCI monomer suggest that free heavy chain cis interactions with other surface molecules can promote neuritogenesis while tripartite MHCI interactions with classical MHCI receptors can inhibit axon outgrowth. Together with the results of others, MHCI appears to differentially modulate neuritogenesis and synaptogenesis.


Subject(s)
Cell Polarity/physiology , Histocompatibility Antigens Class I/metabolism , Neurogenesis/physiology , Neurons/physiology , Actins/metabolism , Analysis of Variance , Animals , Axons/physiology , Cell Polarity/drug effects , Cells, Cultured , Embryo, Mammalian , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/physiology , Hippocampus/cytology , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/pharmacology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microtubule-Associated Proteins/metabolism , Neurites/drug effects , Neurites/physiology , Neurons/drug effects , Phosphopyruvate Hydratase/genetics , Recombinant Proteins/pharmacology , Time Factors , Tubulin/metabolism , beta 2-Microglobulin/deficiency
18.
PLoS One ; 6(9): e25337, 2011.
Article in English | MEDLINE | ID: mdl-21966502

ABSTRACT

Antigen-based therapies (ABTs) very effectively prevent the development of type 1 diabetes (T1D) when given to young nonobese diabetic (NOD) mice, however, they have little or no ability to reverse hyperglycemia in newly diabetic NOD mice. More importantly, ABTs have not yet demonstrated an ability to effectively preserve residual ß-cells in individuals newly diagnosed with type 1 diabetes (T1D). Accordingly, there is great interest in identifying new treatments that can be combined with ABTs to safely protect ß-cells in diabetic animals. The activation of γ-aminobutyric acid (GABA) receptors (GABA-Rs) on immune cells has been shown to prevent T1D, experimental autoimmune encephalomyelitis (EAE) and rheumatoid arthritis in mouse models. Based on GABA's ability to inhibit different autoimmune diseases and its safety profile, we tested whether the combination of ABT with GABA treatment could prolong the survival of transplanted ß-cells in newly diabetic NOD mice. Newly diabetic NOD mice were untreated, or given GAD/alum (20 or 100 µg) and placed on plain drinking water, or water containing GABA (2 or 6 mg/ml). Twenty-eight days later, they received syngenic pancreas grafts and were monitored for the recurrence of hyperglycemia. Hyperglycemia reoccurred in the recipients given plain water, GAD monotherapy, GABA monotherapy, GAD (20 µg)+GABA (2 mg/ml), GAD (20 µg)+GABA (6 mg/ml) and GAD (100 µg)+GABA (6 mg/ml) about 1, 2-3, 3, 2-3, 3-8 and 10-11 weeks post-transplantation, respectively. Thus, combined GABA and ABT treatment had a synergistic effect in a dose-dependent fashion. These findings suggest that co-treatment with GABA (or other GABA-R agonists) may provide a new strategy to safely enhance the efficacy of other therapeutics designed to prevent or reverse T1D, as well as other T cell-mediated autoimmune diseases.


Subject(s)
Diabetes Mellitus, Type 1/prevention & control , Diabetes Mellitus, Type 1/therapy , Islets of Langerhans Transplantation , gamma-Aminobutyric Acid/therapeutic use , Animals , Diabetes Mellitus, Type 1/drug therapy , Female , Glutamate Decarboxylase/therapeutic use , Humans , Hyperglycemia/drug therapy , Hyperglycemia/therapy , Mice , Mice, Inbred NOD
19.
PLoS One ; 6(9): e25338, 2011.
Article in English | MEDLINE | ID: mdl-21966503

ABSTRACT

Adipocyte and ß-cell dysfunction and macrophage-related chronic inflammation are critical for the development of obesity-related insulin resistance and type 2 diabetes mellitus (T2DM), which can be negatively regulated by Tregs. Our previous studies and those of others have shown that activation of γ-aminobutyric acid (GABA) receptors inhibits inflammation in mice. However, whether GABA could modulate high fat diet (HFD)-induced obesity, glucose intolerance and insulin resistance has not been explored. Here, we show that although oral treatment with GABA does not affect water and food consumption it inhibits the HFD-induced gain in body weights in C57BL/6 mice. Furthermore, oral treatment with GABA significantly reduced the concentrations of fasting blood glucose, and improved glucose tolerance and insulin sensitivity in the HFD-fed mice. More importantly, after the onset of obesity and T2DM, oral treatment with GABA inhibited the continual HFD-induced gain in body weights, reduced the concentrations of fasting blood glucose and improved glucose tolerance and insulin sensitivity in mice. In addition, oral treatment with GABA reduced the epididymal fat mass, adipocyte size, and the frequency of macrophage infiltrates in the adipose tissues of HFD-fed mice. Notably, oral treatment with GABA significantly increased the frequency of CD4(+)Foxp3(+) Tregs in mice. Collectively, our data indicated that activation of peripheral GABA receptors inhibited the HFD-induced glucose intolerance, insulin resistance, and obesity by inhibiting obesity-related inflammation and up-regulating Treg responses in vivo. Given that GABA is safe for human consumption, activators of GABA receptors may be valuable for the prevention of obesity and intervention of T2DM in the clinic.


Subject(s)
Diet, High-Fat/adverse effects , Glucose Intolerance/drug therapy , Inflammation/drug therapy , Insulin Resistance/physiology , gamma-Aminobutyric Acid/therapeutic use , Administration, Oral , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Flow Cytometry , Forkhead Transcription Factors/metabolism , Glucose Intolerance/metabolism , Inflammation/chemically induced , Inflammation/metabolism , Intra-Abdominal Fat/metabolism , Mice , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/metabolism , gamma-Aminobutyric Acid/administration & dosage
20.
Autoimmunity ; 44(6): 465-70, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21604972

ABSTRACT

Current treatments for rheumatoid arthritis (RA) have long-term side effects such that new treatments are needed that can safely help manage the disease. There is a growing appreciation that GABA receptors (GABA-Rs) on immune cells provide new targets that can be used to modulate immune cell activity. Here, we show for the first time that activation of peripheral GABA-Rs can inhibit the development of disease in the collagen-induced arthritis (CIA) mouse model of RA. Mice that received oral GABA had a reduced incidence of CIA, and those mice that did develop CIA had milder symptoms. T cells from GABA-treated mice displayed reduced proliferative responses to collagen and their APC had a reduced ability to promote the proliferation of collagen-reactive T cells. Thus, GABA downregulated both T-cell autoimmunity and APC activity. Collagen-reactive T cells from GABA-treated mice displayed reduced recall responses in the presence of GABA ex vivo, indicating that GABA consumption did not desensitize these cells to GABA. GABA-treated mice had reduced collagen-reactive IgG2a, but not IgG1 antibodies, consistent with reduced Th1 help. The levels of serum anti-collagen IgG2a antibodies were correlated significantly with the CIA disease scores of individual mice. Our results suggest that activation of peripheral GABA-Rs may provide a new modality to modulate T cell, B cell, and APC activity and help ameliorate RA and other inflammatory diseases.


Subject(s)
Arthritis, Experimental/drug therapy , Arthritis, Experimental/immunology , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/immunology , Receptors, GABA/immunology , gamma-Aminobutyric Acid/therapeutic use , Animals , Arthritis, Rheumatoid/chemically induced , Collagen , Immunoglobulin G/blood , Lymphocyte Activation/drug effects , Mice , Mice, Inbred DBA , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , gamma-Aminobutyric Acid/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL