Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 86
Filter
1.
Front Physiol ; 13: 1059686, 2022.
Article in English | MEDLINE | ID: mdl-36620212

ABSTRACT

Introduction:Neutrophil transmigration is multifactorial and primarily driven by selectins and ß2-integrins (CD11b/CD18), whose expression are dependent on the underlying stimulus. Ventilator-induced lung injury (VILI) results in a predominantly CD18-independent mechanism of neutrophil recruitment, while direct endotoxin-induced lung injury results from a CD18-dependent mechanism. We previously observed that lack of NADPH oxidases DUOX1 and DUOX2 resulted in reduced neutrophil influx in a VILI model of lung injury but had no influence on neutrophil influx after LPS exposure. Based on these observations, we hypothesized that DUOX1/DUOX2 are an important component of CD18-independent mechanisms of neutrophil recruitment in the lung. Methods:We exposed Duoxa -/- (KO) mice and Duoxa +/+ (WT) mice to either an intratracheal exposure of lipopolysaccharide (LPS/endotoxin)-or high tidal volume ventilation and compared expression of neutrophil markers between groups. WT mice (129S6/SvEvTac) were obtained from Taconic Biosciences (One Discovery Drive Suite 304; Rensselaer, NY 1244) and were allowed to acclimatize for one week prior to study enrollment. KO mice were generated as previously described [Grasberger 2012] and bred in-house on a 129S6 background. We provided positive-pressure ventilation at a tidal volume of 10 ml/kg with 2 cmH20 positive end-expiratory pressure (PEEP). Mice were assigned to groups consisting of KO (n = 5) and WT (n = 5) in each group and divided into non-ventilated, positive-pressure ventilation, or LPS IT exposure groups. Positive-pressure ventilation was instituted for 4-h using a FlexiVent (Flexiware 8.1, Scireq, Montreal, QC, Canada). Lipopolysaccharide (Salmonella enterica serotype tryphimurium L6143, Millipore Sigma) was administered via an intratracheal (IT) route at a dose of 0.1 mg/kg. Mice were humanely euthanized at 4-h post-injection consistent with the UC Davis IAUCAC-approved protocol. Results:As previously observed, neutrophilic influx into the airways was significantly impaired in the Duoxa -/- (KO) mice after VILI, but not after LPS exposure. LPS-induced lung injury resulted in upregulation of CD11b+ neutrophils and shedding of CD62L and CD162 regardless of DUOX expression, whereas VILI resulted in upregulation of CD49+ neutrophils in the Duoxa +/+ (WT) mice but not the Duoxa -/- (KO) mice. Conclusion:Our data suggest DUOX is required for CD18-independent mechanisms of neutrophil recruitment in the lung induced by acute lung injury, but not for canonical CD18depedent mechanisms after LPS exposure.

2.
Eur Rev Med Pharmacol Sci ; 24(16): 8551-8565, 2020 08.
Article in English | MEDLINE | ID: mdl-32894560

ABSTRACT

OBJECTIVE: Our goal was to assess the efficacy of encapsulated allogeneic islets transplanted in diabetic NOD mice and streptozotocin (STZ)-diabetic nonhuman primates (NHPs). MATERIALS AND METHODS: Murine or NHP islets were microencapsulated and transplanted in non-immunosuppressed mice or NHPs given clinically-acceptable immunosuppressive regimens, respectively. Two NHPs were treated with autologous mesenchymal stem cells (MSCs) and peri-transplant oxygen therapy. Different transplant sites (intraperitoneal [i.p.], omental pouch, omental surface, and bursa omentalis) were tested in separate NHPs. Graft function was monitored by exogenous insulin requirements, fasting blood glucose levels, glucose tolerance tests, percent hemoglobin A1c (% HbA1c), and C-peptide levels. In vitro assessment of grafts included histology, immunohistochemistry, and viability staining; host immune responses were characterized by flow cytometry and cytokine/chemokine multiplex ELISAS. RESULTS: Microencapsulated islet allografts functioned long-term i.p. in diabetic NOD mice without immunosuppression, but for a relatively short time in immunosuppressed NHPs. In the NHPs, encapsulated allo-islets initially reduced hyperglycemia, decreased exogenous insulin requirements, elevated C-peptide levels, and lowered % HbA1c in plasma, but graft function diminished with time, regardless of transplant site. At necropsy, microcapsules were intact and non-fibrotic, but many islets exhibited volume loss, central necrosis and endogenous markers of hypoxia. Animals receiving supplemental oxygen and autologous MSCs showed improved graft function for a longer post-transplant period. In diabetic NHPs and mice, cell-free microcapsules did not elicit a fibrotic response. CONCLUSIONS: The evidence suggested that hypoxia was a major factor for damage to encapsulated islets in vivo. To achieve long-term function, new approaches must be developed to increase the oxygen supply to microencapsulated islets and/or identify donor insulin-secreting cells which can tolerate hypoxia.


Subject(s)
Allografts , Diabetes Mellitus, Experimental/therapy , Islets of Langerhans Transplantation , Animals , Capsules/chemistry , Mice , Mice, Inbred NOD
3.
Article in English | MEDLINE | ID: mdl-34164564

ABSTRACT

The coronavirus SARS-CoV-2 is cause of a global pandemic of a pneumonia-like disease termed Coronavirus Disease 2019 (COVID-19). COVID-19 presents a high mortality rate, estimated at 3.4%. More than 1 out of 4 hospitalized COVID-19 patients require admission to an Intensive Care Unit (ICU) for respiratory support, and a large proportion of these ICU-COVID-19 patients, between 17% and 46%, have died. In these patients COVID-19 infection causes an inflammatory response in the lungs that can progress to inflammation with cytokine storm, Acute Lung Injury (ALI), Acute Respiratory Distress Syndrome (ARDS), thromboembolic events, disseminated intravascular coagulation, organ failure, and death. Mesenchymal Stem Cells (MSCs) are potent immunomodulatory cells that recognize sites of injury, limit effector T cell reactions, and positively modulate regulatory cell populations. MSCs also stimulate local tissue regeneration via paracrine effects inducing angiogenic, anti-fibrotic and remodeling responses. MSCs can be derived in large number from the Umbilical Cord (UC). UC-MSCs, utilized in the allogeneic setting, have demonstrated safety and efficacy in clinical trials for a number of disease conditions including inflammatory and immune-based diseases. UC-MSCs have been shown to inhibit inflammation and fibrosis in the lungs and have been utilized to treat patients with severe COVID-19 in pilot, uncontrolled clinical trials, that reported promising results. UC-MSCs processed at our facility have been authorized by the FDA for clinical trials in patients with an Alzheimer's Disease, and in patients with Type 1 Diabetes (T1D). We hypothesize that UC-MSC will also exert beneficial therapeutic effects in COVID-19 patients with cytokine storm and ARDS. We propose an early phase controlled, randomized clinical trial in COVID-19 patients with ALI/ARDS. Subjects in the treatment group will be treated with two doses of UC-MSC (l00 × 106 cells). The first dose will be infused within 24 hours following study enrollment. A second dose will be administered 72 ± 6 hours after the first infusion. Subject in the control group will receive infusion of vehicle (DPBS supplemented with 1% HSA and 70 U/kg unfractionated Heparin, delivered IV) following the same timeline. Subjects will be evaluated daily during the first 6 days, then at 14, 28, 60, and 90 days following enrollment (see Schedule of Assessment for time window details). Safety will be determined by adverse events (AEs) and serious adverse events (SAEs) during the follow-up period. Efficacy will be defined by clinical outcomes, as well as a variety of pulmonary, biochemical and immunological tests. Success of the current study will provide a framework for larger controlled, randomized clinical trials and a means of accelerating a possible solution for this urgent but unmet medical need. The proposed early phase clinical trial will be performed at the University of Miami (UM), in the facilities of the Diabetes Research Institute (DRI), UHealth Intensive Care Unit (ICU) and the Clinical Translational Research Site (CTRS) at the University of Miami Miller School of Medicine and at the Jackson Memorial Hospital (JMH).

4.
Ultrasound Obstet Gynecol ; 54(5): 643-649, 2019 Nov.
Article in English | MEDLINE | ID: mdl-30779235

ABSTRACT

OBJECTIVES: To evaluate fetal growth in pregnancies complicated by placenta previa with or without placenta accreta spectrum (PAS) disorder, compared with in pregnancies with a low-lying placenta. METHODS: This was a multicenter retrospective cohort study of singleton pregnancies complicated by placenta previa with or without PAS disorder, for which maternal characteristics, ultrasound-estimated fetal weight and birth weight were available. Four maternal-fetal medicine units participated in data collection of diagnosis, treatment and outcome. The control group comprised singleton pregnancies with a low-lying placenta (0.5-2 cm from the internal os). The diagnosis of PAS and depth of invasion were confirmed at delivery using both a predefined clinical grading score and histopathological examination. For comparison of pregnancy characteristics and fetal growth parameters, the study groups were matched for smoking status, ethnic origin, fetal sex and gestational age at delivery. RESULTS: The study included 82 women with placenta previa with PAS disorder, subdivided into adherent (n = 35) and invasive (n = 47) PAS subgroups, and 146 women with placenta previa without PAS disorder. There were 64 controls with a low-lying placenta. There was no significant difference in the incidence of small-for-gestational age (SGA) (birth weight ≤ 10th percentile) and large-for-gestational age (LGA) (birth weight ≥ 90th  percentile) between the study groups. Median gestational age at diagnosis was significantly lower in pregnancies with placenta previa without PAS disorder than in the low-lying placenta group (P = 0.002). No significant difference was found between pregnancies complicated by placenta previa with PAS disorder and those without for any of the variables. Median estimated fetal weight percentile was significantly lower in the adherent compared with the invasive previa-PAS subgroup (P = 0.047). Actual birth weight percentile at delivery did not differ significantly between the subgroups (P = 0.804). CONCLUSIONS: No difference was seen in fetal growth in pregnancies complicated by placenta previa with PAS disorder compared with those without and compared with those with a low-lying placenta. There was also no increased incidence of either SGA or LGA neonates in pregnancies with placenta previa and PAS disorder compared with those with placenta previa with spontaneous separation of the placenta at birth. Adverse neonatal outcome in pregnancies complicated by placenta previa and PAS disorder is linked to premature delivery and not to impaired fetal growth. Copyright © 2019 ISUOG. Published by John Wiley & Sons Ltd.


Subject(s)
Birth Weight , Fetal Development , Placenta Accreta/physiopathology , Placenta Previa/physiopathology , Adult , Case-Control Studies , Female , Humans , Infant, Newborn , Placenta/pathology , Pregnancy , Retrospective Studies , Ultrasonography, Prenatal
5.
Allergy ; 71(1): 5-14, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26335713

ABSTRACT

The study of autophagy ('self-eating'), a fundamental cell fate pathway involved in physiological and pathological subcellular processes, opens a new frontier in the continuous search for novel therapies for human asthma. Asthma is a complex syndrome with different disease phenotypes. Autophagy plays a central role in cell physiology, energy and metabolism, and cell survival. Autophagy's hallmark is the formation of double-membrane autophagic autophagosomes, and this process is operational in airway epithelial and mesenchymal cells in asthma. Genetic associations between autophagy genes and asthma have been observed including single nucleotide polymorphisms in Atg5 which correlate with reduced lung function. Immune mechanisms important in asthma such as Th2 cells and eosinophils also manifest autophagy. Lastly, we address the role of autophagy in extracellular matrix deposition and fibrosis in asthmatic airways remodeling, a pathologic process still without effective therapy, and discuss potential pharmacologic inhibitors. We end by offering two opposing but plausible hypotheses as to how autophagy may be directly involved in airway fibrosis.


Subject(s)
Asthma/etiology , Asthma/metabolism , Autophagy , Airway Remodeling , Animals , Anti-Asthmatic Agents/pharmacology , Anti-Asthmatic Agents/therapeutic use , Asthma/drug therapy , Asthma/pathology , Autophagy/drug effects , Autophagy/genetics , Autophagy/immunology , Disease Models, Animal , Extracellular Matrix/immunology , Extracellular Matrix/metabolism , Fibrosis , Genetic Predisposition to Disease , Humans , Immunity
6.
Clin Rev Allergy Immunol ; 48(1): 7-30, 2015 Feb.
Article in English | MEDLINE | ID: mdl-24178860

ABSTRACT

The critical asthma syndrome (CAS) encompasses the most severe, persistent, refractory asthma patients for the clinician to manage. Personalized pharmacotherapy is necessary to prevent the next acute severe asthma exacerbation, not just the control of symptoms. The 2007 National Asthma Education and Prevention Program Expert Panel 3 provides guidelines for the treatment of uncontrolled asthma. The patient's response to recommended pharmacotherapy is highly variable which risks poor asthma control leading to frequent exacerbations that can deteriorate into CAS. Controlling asthma symptoms and preventing acute exacerbations may be two separate clinical activities with their own unique demands. Clinicians must be prepared to use the entire spectrum of asthma medications available but must concurrently be aware of potential drug toxicities some of which can paradoxically worsen asthma control. Medications normally prescribed for COPD can potentially be useful in the CAS patient, particularly those with asthma-COPD overlap syndrome. Immunomodulation with drugs like omalizumab in IgE-mediated asthma syndromes is one important approach. New and emerging drugs address unique aspects of airway inflammation and biology but at a significant financial cost. The pharmacology and toxicities of the agents that may be used in the treatment of CAS to control asthma symptoms and prevent severe exacerbations are reviewed.


Subject(s)
Anti-Asthmatic Agents/therapeutic use , Antibodies, Anti-Idiotypic/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Asthma/drug therapy , Pulmonary Disease, Chronic Obstructive/drug therapy , Respiratory System/drug effects , Animals , Critical Illness , Drug Therapy/trends , Expert Testimony , Humans , Immunoglobulin E/immunology , Immunoglobulin E/metabolism , Immunomodulation , Omalizumab , Practice Guidelines as Topic , Respiratory System/immunology , Syndrome
7.
Clin Exp Immunol ; 170(2): 131-8, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23039883

ABSTRACT

The mRNA levels of a set of immune-related genes were analysed with peripheral blood samples from at-risk, new-onset and long-term type 1 diabetes (T1D) patients, in comparison to those from healthy controls. The selected set includes T lymphocyte genes [CD3G and cytotoxic T lymphocyte-associated antigen 4 (CTLA4)], B lymphocyte genes (CD19 and CD20) and myeloid cell-related genes [CD11b, Toll-like receptor (TLR)-9, arginase (ARG1)]. Also included is a subset of the S100 family members that has been documented recently as regulatory elements of innate immunity. Samples from patients with long-term T1D had a reduced level of mRNA for most of selected innate and adaptive immune genes. No such reduction was detected in samples collected from at-risk or new-onset T1D patients. Analyses of regulatory gene expression ratios revealed a dynamic disproportion of CTLA4 versus CD3G expression in samples from at-risk, new-onset and long-term T1D patients. These changes could serve as immunological biomarkers for the status of the immune system during T1D progression and therapeutic interventions.


Subject(s)
Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Adaptive Immunity/genetics , Adaptive Immunity/immunology , Adolescent , Adult , Antigens, CD/blood , Antigens, CD/genetics , Antigens, CD/immunology , Antigens, CD/metabolism , Arginase/blood , Arginase/genetics , Arginase/immunology , Arginase/metabolism , Biomarkers/blood , CTLA-4 Antigen/blood , CTLA-4 Antigen/genetics , CTLA-4 Antigen/immunology , CTLA-4 Antigen/metabolism , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/metabolism , Female , Gene Expression Profiling/methods , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Humans , Immunity, Innate/genetics , Immunity, Innate/immunology , Male , Middle Aged , RNA, Messenger/genetics , RNA, Messenger/immunology , S100 Proteins/blood , S100 Proteins/genetics , S100 Proteins/immunology , S100 Proteins/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Toll-Like Receptor 9/blood , Toll-Like Receptor 9/genetics , Toll-Like Receptor 9/immunology , Toll-Like Receptor 9/metabolism , Young Adult
8.
Diabetologia ; 54(5): 1121-6, 2011 May.
Article in English | MEDLINE | ID: mdl-21360190

ABSTRACT

AIMS/HYPOTHESIS: The aim of this study was to provide evidence that the anterior chamber of the eye serves as a novel clinical islet implantation site. METHODS: In a preclinical model, allogeneic pancreatic islets were transplanted into the anterior chamber of the eye of a baboon model for diabetes, and metabolic and ophthalmological outcomes were assessed. RESULTS: Islets readily engrafted on the iris and there was a decrease in exogenous insulin requirements due to insulin secretion from the intraocular grafts. No major adverse effects on eye structure and function could be observed during the transplantation period. CONCLUSIONS/INTERPRETATION: Our study demonstrates the long-term survival and function of allogeneic islets after transplantation into the anterior chamber of the eye. The safety and simplicity of this procedure provides support for further studies aimed at translating this technology into the clinic.


Subject(s)
Anterior Chamber/surgery , Diabetes Mellitus, Experimental/therapy , Islets of Langerhans Transplantation/methods , Animals , Papio
9.
Pharmazie ; 65(6): 421-8, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20614690

ABSTRACT

Emerging biotechnologies, such as the use of biohybrid devices for cellular therapies, are showing increasing therapeutic promise for the treatment of various diseases, including type 1 diabetes mellitus. The functionality of such devices could be greatly enhanced if successful localized immunosuppression regimens could be established, since they would eliminate the many otherwise unavoidable side effects of currently used systemic immunosuppressive therapies. The existence of local immune privilege at some specialized tissues, such as the eye, CNS, or pregnant uterus, supports the feasibility of localized immunomodulation, and such an approach is particularly well-suited for cell transplant therapies where all transplanted tissue is localized within a device. Following the success of syngeneic transplantation in a subcutaneous prevascularized device as a bioartificial pancreas in a rodent model, we now report the first results of exploratory in vivo islet allograft studies in rats using locally delivered glucocorticoids (dexamethasone phosphate and the soft steroid loteprednol etabonate). Following in vitro assessments, in silico drug distribution models were used to establish tentative therapeutic dose ranges. Sustained local delivery was achieved via implantable osmotic mini-pumps through a central sprinkler, as well as with a sustained-delivery formulation for loteprednol etabonate using poly(D,L-lactic) acid (PLA) microspheres. Doses delivered locally were approximately hundred-fold smaller than those typically used in systemic treatments. While several solubility, stability, and implantation problems still remain to be addressed, both compounds showed promise in their ability to prolong graft survival after tapering of systemic immunosuppression, compared to control groups.


Subject(s)
Cell Transplantation/instrumentation , Glucocorticoids/pharmacology , Immunosuppressive Agents , Islets of Langerhans Transplantation/immunology , Algorithms , Androstadienes/administration & dosage , Animals , Biotechnology , Computer Simulation , Delayed-Action Preparations , Drug Delivery Systems , Drug Implants , Feasibility Studies , Female , Glucocorticoids/administration & dosage , Glucocorticoids/adverse effects , Humans , Immunosuppressive Agents/adverse effects , Kaplan-Meier Estimate , Lactic Acid , Loteprednol Etabonate , Microspheres , Polyglycolic Acid , Polylactic Acid-Polyglycolic Acid Copolymer , Pregnancy , Rats , Tissue Distribution
10.
Am J Transplant ; 9(1): 91-104, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19133931

ABSTRACT

The aim of this study was to test whether an omental pouch can be used as an alternative site for islet implantation in diabetic monkeys. Here we report the successful engraftment of islets in diabetic cynomolgus monkeys when loaded on a synthetic biodegradable scaffold and placed in an omental pouch. One autologous and five allogeneic diabetic monkey transplants under the cover of steroid-free immune suppression (SFIS) were undertaken. Fasting blood glucose (FBG) and C-peptide (CP), exogenous insulin requirements (EIR), intravenous glucose tolerance test (IVGTT), A1C and histopathology were used to assess islet engraftment and survival. All animals achieved CP levels > 1.0 ng/mL following transplant, a 66-92% posttransplant decrease in EIR and reduced A1C. Following graft removal, CP became negative and histopathological analysis of the explanted grafts demonstrated well-granulated and well-vascularized, insulin-positive islets, surrounded by T-cell subsets and macrophages. Compared to intrahepatic allogeneic islet transplants (n = 20), there was a delayed engraftment for omental pouch recipients but similar levels of CP production were ultimately achieved, with a broad range of IEQ/kg transplanted in both sites. Our results suggest this extrahepatic transplantation site has potential as an alternative site for clinical islet cell transplantation.


Subject(s)
Diabetes Mellitus, Experimental/surgery , Graft Survival , Islets of Langerhans Transplantation , Omentum , Animals , Macaca fascicularis , Streptozocin
11.
Am J Transplant ; 8(6): 1262-74, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18444924

ABSTRACT

To prevent graft rejection and avoid immunosuppression-related side-effects, we attempted to induce recipient chimerism and graft tolerance in islet transplantation by donor CD34+hematopoietic stem cell (HSC) infusion. Six patients with brittle type 1 Diabetes Mellitus received a single-donor allogeneic islet transplant (8611 +/- 2113 IEQ/kg) followed by high doses of donor HSC (4.3 +/- 1.9 x 10(6) HSC/kg), at days 5 and 11 posttransplant, without ablative conditioning. An 'Edmonton-like' immunosuppression was administered, with a single dose of anti-TNFalpha antibody (Infliximab) added to induction. Immunosuppression was weaned per protocol starting 12 months posttransplant. After transplantation, glucose control significantly improved, with 3 recipients achieving insulin-independence for a short time (24 +/- 23 days). No severe hypoglycemia or protocol-related adverse events occurred. Graft function was maximal at 3 months then declined. Two recipients rejected within 6 months due to low immunosuppressive trough levels, whereas 4 completed 1-year follow-up with functioning grafts. Graft failure occurred within 4 months from weaning (478 +/- 25 days posttransplant). Peripheral chimerism, as donor leukocytes, was maximal at 1-month (5.92 +/- 0.48%), highly reduced at 1-year (0.20 +/- 0.08%), and was undetectable at graft failure. CD25+T-lymphocytes significantly decreased at 3 months, but partially recovered thereafter. Combined islet and HSC allotransplantation using an 'Edmonton-like' immunosuppression, without ablative conditioning, did not lead to stable chimerism and graft tolerance.


Subject(s)
Chimerism , Diabetes Mellitus, Type 1/surgery , Hematopoietic Stem Cell Transplantation , Islets of Langerhans Transplantation/immunology , Transplantation Tolerance/immunology , Adult , Antigens, CD34/immunology , Female , Humans , Male , Middle Aged , Pilot Projects , Prospective Studies , Transplantation, Homologous
12.
Curr Med Chem ; 14(20): 2204-13, 2007.
Article in English | MEDLINE | ID: mdl-17691958

ABSTRACT

New clinical practice guidelines for patients with asthma include the recommendation to monitor exhaled breath nitric oxide (NO) levels. NO concentrations in exhaled breath are increased in asthmatics and increased NO levels correlate with worsening airway inflammation and asthma symptoms. The multiple roles of NO in the lung have not been delineated clearly. Clinical trials are being performed presently that test the apparently conflicting hypotheses that either donors or inhibitors of NO in the lung are effective strategies for treating asthma. These strategies evolved, in part, from results of pre-clinical studies performed in mice and other animal models. This review evaluates the existing literature with regard to mouse models of asthma and explores the often conflicting data on the role of NO, the nitric oxide synthase (NOS) enzymes, and the arginase enzymes in allergic airway inflammation. While we will emphasize the ovalbumin exposure mouse model, we will also examine other models. Where inconsistencies are identified among the studies, we attempt to determine whether such inconsistencies arise from methodological differences or alternative mechanisms. Ultimately, we address whether the allergen-exposed mouse is a suitable model for identifying promising new drugs for the treatment of human asthma. While a consensus is building that NO is beneficial or protective in subsets of asthmatics, results from studies using mouse models to investigate the individual roles of NO and the NOS enzymes in airway inflammation are often contradictory. Further research efforts with this model will allow us to distinguish which asthma patients may benefit best from NO donors and which may benefit from NO inhibitors.


Subject(s)
Anti-Asthmatic Agents/therapeutic use , Asthma/etiology , Disease Models, Animal , Mice , Nitric Oxide/metabolism , Animals , Anti-Asthmatic Agents/chemistry , Anti-Asthmatic Agents/pharmacology , Asthma/drug therapy , Asthma/metabolism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Nitric Oxide/antagonists & inhibitors , Nitric Oxide Donors/chemistry , Nitric Oxide Donors/pharmacology , Nitric Oxide Donors/therapeutic use , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide Synthase Type II/metabolism
13.
Inflamm Res ; 54(2): 57-65, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15750712

ABSTRACT

OBJECTIVE AND DESIGN: We examined the reversibility of several changes in the lungs and airways of mice immediately after exposure to ovalbumin aerosol and after a period of recovery breathing clean air. METHODS: Mice were exposed for 1, 2, 4, 6, 8, or 10 weeks, with recovery in clean air for 1-3 weeks. RESULTS: Airway collagen content, exhaled NO, airway mucous cell hyperplasia, and lung lavage inflammatory cell content increased upon exposure to ovalbumin aerosol. All parameters except airway fibrosis decreased partially or completely to control values with recovery in clean air. CONCLUSIONS: Airway mucous cell hypertrophy and hyperplasia appear to be completely reversible after recovery in clean air, while exhaled NO and airway inflammation appear to be mostly reversible, except for persistence of lymphocytes in the lung lavage fluid. Airway fibrosis appears to be reversible when mice are exposed to ovalbumin aerosol for periods of up to 4 weeks of exposure, but becomes irreversible after 6 or more weeks of exposure.


Subject(s)
Bronchial Diseases/pathology , Bronchitis/pathology , Ovalbumin/administration & dosage , Pneumonia/pathology , Pulmonary Fibrosis/pathology , Administration, Inhalation , Animals , Bronchial Diseases/chemically induced , Bronchial Diseases/physiopathology , Bronchitis/chemically induced , Bronchitis/physiopathology , Collagen/metabolism , Drug Administration Schedule , Exhalation , Female , Fibrosis , Hyperplasia , Hypertrophy , Male , Mice , Mice, Inbred BALB C , Nitric Oxide , Pneumonia/chemically induced , Pneumonia/physiopathology , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/physiopathology , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology
14.
Thorax ; 58(9): 772-7, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12947136

ABSTRACT

BACKGROUND: Subepithelial collagen and extracellular matrix protein deposition are important pathophysiological components of airway remodelling in chronic asthma. Animal models based on the local reaction to antigens show structural alterations in the airway submucosal region and provide important information regarding disease pathophysiology. We describe a murine model of peribronchial fibrosis using intratracheally instilled transforming growth factor (TGF)-beta(1) in BALB/C mice that facilitates a mechanistic approach to understanding the cellular and molecular pathways leading to airway fibrosis. METHODS: BALB/C mice were intratracheally instilled with either TGF-beta(1) or buffered saline. Airway fibrosis was assessed by light microscopy, hydroxyproline content, and polymerase chain reaction (PCR) for collagen I and III on microdissected airway samples. The lysyl oxidase inhibitor beta-aminoproprionitrile (BAPN) was administered to TGF-beta(1) treated mice to block airway collagen deposition. Airway hyperresponsiveness was also measured after treatment with TGF-beta(1). RESULTS: During the 7 days after administration of TGF-beta(1) the mice developed increased subepithelial collagen which could be blocked by BAPN. Increased mRNAs for collagen types I and III were seen in microdissected airways 1 week after TGF-beta(1), and significantly increased total collagen was found in the airways 4 weeks after TGF-beta(1). A detectable increase in airway hyperreactivity occurred. CONCLUSIONS: This new model should facilitate detailed study of airway remodelling that occurs in the absence of detectable cellular inflammation, and allow examination of the functional consequences of a major structural alteration in the conducting airways uncomplicated by inflammatory cell influx.


Subject(s)
Bronchial Diseases/etiology , Collagen Type III/metabolism , Collagen Type I/metabolism , Pulmonary Fibrosis/etiology , RNA, Messenger/metabolism , Transforming Growth Factor beta/metabolism , Aminopropionitrile/adverse effects , Animals , Bronchi/metabolism , Bronchial Diseases/metabolism , Bronchoalveolar Lavage Fluid/cytology , Immunohistochemistry , Mice , Mice, Inbred BALB C , Pulmonary Fibrosis/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods , Transforming Growth Factor beta1
15.
Diabetes ; 50(8): 1706-13, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11473028

ABSTRACT

The resounding success of a new immunosuppressive regimen known as the Edmonton protocol demonstrates that islet cell transplantation is becoming a therapeutic reality for diabetes. However, under the Edmonton protocol, a single donor does not provide enough islets to attain the insulin independence of a transplant recipient. This limitation is mainly caused by islet apoptosis triggered during isolation. In this study, we describe a highly efficient system of transiently transferring anti-apoptotic proteins into pancreatic islets, thus opening an exciting new therapeutic opportunity to improve the viability of transplantable islets. We fused beta-galactosidase to the 11-amino acid residues that constitute the protein transduction domain (PTD) of the HIV/TAT protein and transduced pancreatic islets ex vivo with this fusion protein in a dose-dependent manner with >80% efficiency. We observed that transduction of the anti-apoptotic proteins Bcl-X(L) and PEA-15 fused to TAT/PTD prevented apoptosis induced by tumor necrosis factor-alpha in a pancreatic beta-cell line, indicating that TAT/PTD anti-apoptotic proteins retained their biological activity. Finally, we demonstrated that TAT-fusion proteins did not affect the insulin secretion capability of islets, as determined by glucose static incubation and by reversion of hyperglycemia in diabetic immunodeficient mice.


Subject(s)
Apoptosis/physiology , Diabetes Mellitus, Experimental/surgery , Gene Products, tat/metabolism , Islets of Langerhans Transplantation/physiology , Islets of Langerhans/cytology , Islets of Langerhans/physiology , Phosphoproteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Transfection/methods , Animals , Apoptosis Regulatory Proteins , Blood Glucose/metabolism , Caspase 3 , Caspases/metabolism , Cell Line , Cells, Cultured , Diabetes Mellitus, Experimental/blood , Gene Products, tat/genetics , HIV/genetics , Hyperglycemia/therapy , Islets of Langerhans Transplantation/pathology , Male , Mice , Mice, SCID , Phosphoproteins/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Rats , Rats, Inbred Lew , Recombinant Fusion Proteins/biosynthesis , Transplantation, Heterologous/physiology , bcl-X Protein , beta-Galactosidase/genetics , tat Gene Products, Human Immunodeficiency Virus
17.
World J Surg ; 25(4): 481-6, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11344402

ABSTRACT

New hope for the treatment of type 1 diabetes has recently emerged from the encouraging results of islet cell transplantation in humans during the last few years. Although still facing considerable problems, the challenge to achieving insulin independence has been overcome in some patients who received an islet graft. However, the success of clinical trials is still limited by the inability to transplant enough viable human islets to compensate for the insulin-deficient state, the number of islets that engraft following transplantation, the rejection process, and the recurrence of autoimmunity. The important advances in immunosuppressive regimens, organ procurement techniques, isolation techniques, and availability of defined collagenase blends have contributed to the continuing promise of making islet cell transplantation the treatment of choice for type 1 diabetes mellitus.


Subject(s)
Diabetes Mellitus, Type 1/surgery , Islets of Langerhans Transplantation , Diabetes Mellitus, Type 1/physiopathology , Graft Rejection , Humans , Islets of Langerhans Transplantation/immunology , Islets of Langerhans Transplantation/physiology , Tissue and Organ Procurement , Transplantation Tolerance
18.
Diabetes ; 50(2): 270-6, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11272136

ABSTRACT

Allorejection and recurrence of autoimmunity are the major barriers to transplantation of islets of Langerhans for the cure of type 1 diabetes in humans. CD40-CD154 (CD40 ligand) interaction blockade by the use of anti-CD154 monoclonal antibody (mAb) has shown efficacy in preventing allorejection in several models of organ and cell transplantation. Here we report the beneficial effect of the chronic administration of a hamster anti-murine CD154 mAb, MR1, in prolonging islet graft survival in NOD mice. We explored the transplantation of C57BL/6 islets into spontaneously diabetic NOD mice, a combination in which both allogeneic and autoimmune components are implicated in graft loss. Recipients were treated either with an irrelevant control antibody or with MR1. MR1 administration was effective in prolonging allograft survival, but did not provide permanent protection from diabetes recurrence. The autoimmune component of graft loss was studied in spontaneously diabetic NOD mice that received syngeneic islets from young male NOD mice. In this combination, a less dramatic yet substantial delay in diabetes recurrence was observed in the MR1-treated recipients when compared with the control group. Finally, the allogeneic component was explored by transplanting C57BL/6 islets into chemically induced diabetic male NOD mice. In this setting, long-term graft survival (>100 days) was achieved in MR1-treated mice, whereas control recipients rejected their grafts within 25 days. In conclusion, chronic blockade of CD154 results in permanent protection from allorejection and significantly delays recurrence of diabetes in NOD mice.


Subject(s)
Antibodies, Monoclonal/pharmacology , CD40 Antigens/immunology , CD40 Ligand/immunology , Islets of Langerhans Transplantation , Mice, Inbred NOD/physiology , T-Lymphocytes/immunology , Animals , CD4-CD8 Ratio , Diabetes Mellitus/genetics , Diabetes Mellitus/surgery , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Experimental/surgery , Graft Survival/drug effects , Immunohistochemistry , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Islets of Langerhans Transplantation/immunology , Male , Mice , Mice, Inbred C57BL , T-Lymphocyte Subsets/drug effects , Time Factors , Transplantation, Homologous
SELECTION OF CITATIONS
SEARCH DETAIL