Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Med Sci Educ ; 33(1): 147-156, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36688011

ABSTRACT

Purpose: Virtual instruction became the primary educational delivery method for pre-clerkship medical students during the COVID-19 pandemic. The aims of this study were to evaluate the effectiveness of a virtual and blended pre-clerkship curriculum and to assess its impact on students. Methods: We surveyed 223 1st- and 2nd-year medical students (MS1s and MS2s) enrolled at the Paul L Foster School of Medicine. We analyzed student satisfaction with their courses, along with summative exam scores, compared to previous academic years. Results: The survey was completed by 125 of 223 students (56%). Most students changed their study methods (78%), experienced technical issues (85%), and had difficulty communicating with faculty (62%). MS1s were significantly more likely than MS2s to report difficulty in adjusting to virtual instruction (p = 0.037) and a negative impact on their learning skills (p = 0.005) and academic performance (p = 0.003). Students reported the virtual environment negatively affected their social skills (77%), connectedness to peers (89%), and professional development (62%). MS1s were more likely than MS2s to perceive a negative effect on their sense of wellness (p = 0.002). The overall satisfaction with the courses was similar to previous academic years. Student performance in the summative examination of the first virtually delivered unit was lower (p = 0.007) than the previous year's cohorts. Conclusion: The difference in MS1s and MS2s perceptions of virtual and blended instruction highlights the importance of face-to-face learning during the first year. Benefits and drawbacks were identified which may help inform educators when designing future learning models. Supplementary Information: The online version contains supplementary material available at 10.1007/s40670-022-01723-6.

2.
Germs ; 11(2): 314-318, 2021 Jun.
Article in English | MEDLINE | ID: mdl-34422702

ABSTRACT

INTRODUCTION: Lemierre's syndrome refers to the septic thrombophlebitis of the internal jugular vein, secondary to a pharyngeal infection. Although it mainly affects the internal jugular vein, isolated cases have been described of involvement of the external jugular vein. The main etiological agent is Fusobacterium necrophorum. CASE REPORT: A 27-year-old male, previously healthy, presented with a 7-day history of sore throat and fever. He was diagnosed with Lemierre's syndrome, coinfection by Bacillus circulans, F. nucleatum and Staphylococcus aureus with an atypical presentation due to the involvement of the external jugular vein and the internal jugular vein. CONCLUSIONS: As far as we are aware, we present the first case of Lemierre's syndrome with these characteristics.

3.
Sci Transl Med ; 13(575)2021 01 06.
Article in English | MEDLINE | ID: mdl-33408187

ABSTRACT

Necrotizing enterocolitis (NEC) causes acute intestinal necrosis in premature infants and is associated with severe neurological impairment. In NEC, Toll-like receptor 4 is activated in the intestinal epithelium, and NEC-associated brain injury is characterized by microglial activation and white matter loss through mechanisms that remain unclear. We now show that the brains of mice and humans with NEC contained CD4+ T lymphocytes that were required for the development of brain injury. Inhibition of T lymphocyte influx into the brains of neonatal mice with NEC reduced inflammation and prevented myelin loss. Adoptive intracerebroventricular delivery of gut T lymphocytes from mice with NEC into Rag1 -/- recipient mice lacking CD4+ T cells resulted in brain injury. Brain organoids derived from mice with or without NEC and from human neuronal progenitor cells revealed that IFN-γ release by CD4+ T lymphocytes induced microglial activation and myelin loss in the organoids. IFN-γ knockdown in CD4+ T cells derived from mice with NEC abrogated the induction of NEC-associated brain injury after adoptive transfer to naïve Rag1 -/- recipient mice. T cell receptor sequencing revealed that NEC mouse brain-derived T lymphocytes shared homology with gut T lymphocytes from NEC mice. Intraperitoneal injection of NEC gut-derived CD4+ T lymphocytes into naïve Rag1 -/- recipient mice induced brain injury, suggesting that gut-derived T lymphocytes could mediate neuroinflammation in NEC. These findings indicate that NEC-associated brain injury may be induced by gut-derived IFN-γ-releasing CD4+ T cells, suggesting that early management of intestinal inflammation in children with NEC could improve neurological outcomes.


Subject(s)
Enterocolitis, Necrotizing , Animals , Brain , Inflammation , Intestinal Mucosa , Mice , T-Lymphocytes, Regulatory
5.
Pediatr Res ; 89(1): 91-101, 2021 01.
Article in English | MEDLINE | ID: mdl-32221473

ABSTRACT

BACKGROUND: Necrotizing enterocolitis (NEC) develops through exaggerated toll-like receptor 4 (TLR4) signaling in the intestinal epithelium. Breast milk is rich in non-digestible oligosaccharides and prevents NEC through unclear mechanisms. We now hypothesize that the human milk oligosaccharides 2'-fucosyllactose (2'-FL) and 6'-sialyllactose (6'-SL) can reduce NEC through inhibition of TLR4 signaling. METHODS: NEC was induced in newborn mice and premature piglets and infant formula was supplemented with 2'-FL, 6'-SL, or lactose. Intestinal tissue was obtained at surgical resection. HMO inhibition of TLR4 was assessed in IEC-6 enterocytes, mice, and human tissue explants and via in silico modeling. RESULTS: Supplementation of infant formula with either 2'-FL and/or 6'-SL, but not the parent sugar lactose, reduced NEC in mice and piglets via reduced apoptosis, inflammation, weight loss, and histological appearance. Mechanistically, both 2'-FL and 6'-SL, but not lactose, reduced TLR4-mediated nuclear factor kappa light-chain enhancer of activated B cells (NF-kB) inflammatory signaling in the mouse and human intestine. Strikingly, in silico modeling revealed 2'-FL and 6'-SL, but not lactose, to dock into the binding pocket of the TLR4-MD2 complex, explaining their ability to inhibit TLR4 signaling. CONCLUSIONS: 2'-FL and 6'-SL, but not lactose, prevent NEC in mice and piglet models and attenuate NEC inflammation in the human ileum, in part through TLR4 inhibition. IMPACT: Necrotizing enterocolitis (NEC) is a major cause of morbidity and mortality in premature infants that occurs in the setting of bacterial colonization of the gut and administration of formula feeds and activation by the innate immune receptor toll-like receptor 4 (TLR4). Breast milk prevents NEC through unclear mechanisms. We now show that breast milk-enriched human milk oligosaccharides (HMOs) that are derived from lactose prevent NEC through inhibition of TLR4. The human milk oligosaccharides 2'-FL and 6'-SL, but not the backbone sugar lactose, prevent NEC in mice and piglets. 2'-FL and 6'-SL but not lactose inhibited TLR4 signaling in cultured enterocytes, in enteroids derived from mouse intestine, and in human intestinal explants obtained at the time of surgical resection for patients with NEC. In seeking the mechanisms involved, 2'-FL and 6'-SL but not lactose were found to directly bind to TLR4, explaining the inhibition and protection against NEC. These findings may impact clinical practice by suggesting that administration of HMOs could serve as a preventive strategy for premature infants at risk for NEC development.


Subject(s)
Enterocolitis, Necrotizing/prevention & control , Ileum/drug effects , Intestinal Mucosa/drug effects , Lactose/analogs & derivatives , Milk, Human/chemistry , Toll-Like Receptor 4/antagonists & inhibitors , Trisaccharides/pharmacology , Animals , Animals, Newborn , Apoptosis/drug effects , Cell Line , Disease Models, Animal , Enterocolitis, Necrotizing/immunology , Enterocolitis, Necrotizing/metabolism , Enterocolitis, Necrotizing/pathology , Humans , Ileum/immunology , Ileum/metabolism , Ileum/pathology , Inflammation Mediators/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Lactose/isolation & purification , Lactose/pharmacology , Mice , Molecular Docking Simulation , Signal Transduction , Sus scrofa , Toll-Like Receptor 4/metabolism , Trisaccharides/isolation & purification , Weight Loss/drug effects
6.
Sci Transl Med ; 10(471)2018 12 12.
Article in English | MEDLINE | ID: mdl-30541786

ABSTRACT

Necrotizing enterocolitis (NEC) is a severe gastrointestinal disease of the premature infant. One of the most important long-term complications observed in children who survive NEC early in life is the development of profound neurological impairments. However, the pathways leading to NEC-associated neurological impairments remain unknown, thus limiting the development of prevention strategies. We have recently shown that NEC development is dependent on the expression of the lipopolysaccharide receptor Toll-like receptor 4 (TLR4) on the intestinal epithelium, whose activation by bacteria in the newborn gut leads to mucosal inflammation. Here, we hypothesized that damage-induced production of TLR4 endogenous ligands in the intestine might lead to activation of microglial cells in the brain and promote cognitive impairments. We identified a gut-brain signaling axis in an NEC mouse model in which activation of intestinal TLR4 signaling led to release of high-mobility group box 1 in the intestine that, in turn, promoted microglial activation in the brain and neurological dysfunction. We further demonstrated that an orally administered dendrimer-based nanotherapeutic approach to targeting activated microglia could prevent NEC-associated neurological dysfunction in neonatal mice. These findings shed light on the molecular pathways leading to the development of NEC-associated brain injury, provide a rationale for early removal of diseased intestine in NEC, and indicate the potential of targeted therapies that protect the developing brain in the treatment of NEC in early childhood.


Subject(s)
Brain/pathology , Cognitive Dysfunction/etiology , Enterocolitis, Necrotizing/complications , Microglia/pathology , Acetylcysteine/pharmacology , Acetylcysteine/therapeutic use , Administration, Oral , Animals , Animals, Newborn , Antioxidants/administration & dosage , Antioxidants/pharmacology , Antioxidants/therapeutic use , Brain/ultrastructure , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/pathology , Dendrimers/chemistry , HMGB1 Protein/metabolism , Humans , Intestinal Mucosa/metabolism , Mice, Inbred C57BL , Myelin Sheath/drug effects , Myelin Sheath/metabolism , Myelin Sheath/ultrastructure , Toll-Like Receptor 4/metabolism
7.
J Pediatr Surg ; 53(6): 1192-1196, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29602549

ABSTRACT

PURPOSE: The definitive diagnosis of necrotizing enterocolitis (NEC) is typically at an advanced stage, indicating the need for sensitive and noninvasive diagnostic modalities. Near infrared spectroscopy (NIRS) has been utilized to noninvasively measure intraabdominal tissue oxygenation and to diagnose NEC, but specificity is lacking, in part because sensors are limited to a narrow band of the electromagnetic spectrum. Here, we introduce the concept of broadband optical spectroscopy (BOS) as a noninvasive method to characterize NEC. METHODS: NEC was induced in 7-day old mice by gavage feeding with formula supplemented with enteric bacteria plus hypoxia. Transabdominal spectroscopy was performed daily using a broad-spectrum halogen light source coupled with a spectroradiometer capable of detection from 400 to 1800nm. RESULTS: A feature extraction algorithm was developed based on the spectral waveforms from mice with NEC. When subsequently tested on cohorts of diseased and control mice by a blinded examiner, noninvasive BOS was able to detect disease with 100% specificity and sensitivity. CONCLUSIONS: We reveal that the use of BOS is able to accurately and noninvasively discriminate the presence of NEC in a mouse model, thus introducing a noninvasive early diagnostic modality for this devastating disease.


Subject(s)
Enterocolitis, Necrotizing/diagnostic imaging , Optical Imaging/methods , Animals , Early Diagnosis , Mice , Mice, Inbred C57BL , Sensitivity and Specificity
8.
Intensive Care Med Exp ; 5(1): 21, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28405943

ABSTRACT

BACKGROUND: Recognition of cardiomyopathy in sepsis can be challenging due to the limitations of conventional measures such as ejection fraction (EF) and fractional shortening (FS) in the context of variable preload and afterload conditions. This study correlates myocardial function using strain echocardiography (SE) with cardiomyocyte oxidative stress in a murine model of sepsis. METHODS: C57BL/6J mice were randomized into control (n = 10), sham (n = 25), and a cecal ligation and puncture (CLP) (n = 33) model of sepsis. Echocardiography was performed pre-, 12, 24, and 48 h post-injury. Cardiac pro-inflammatory cytokines and mitochondrial redox scavenger expression were evaluated in a subset of each arm. To evaluate the influence of redox scavenger upregulation on oxidative injury and cardiac function, CLP was performed on mitochondrial catalase-upregulated C57BL/6J MCAT+/+ mice (n = 12) and wild-type (WT) animals for comparison. RESULTS: Septic C57BL/6J mice exhibited depressed longitudinal strain (LS) when compared to sham and control at 24 h (p < 0.01) and 48 h (p = 0.04) post-CLP despite having a preserved EF. Furthermore, there was a significant association between increased odds of mortality and depressed LS (OR = 1.23, p = 0.04). Septic C57BL/6J mice concomitantly demonstrated increased expression of cardiomyocyte pro-inflammatory cytokines and decreased expression of redox scavengers at 24 and 48 h. When comparing C57Bl/6 MCAT +/+ mice and C57BL/6J WT mice, a significant decrease in LS was identified in the WT mice at 24 h (MCAT = -23 ± 5% vs. WT = -15 ± 4% p < 0.01) and 48 h (MCAT = -23 ± 7% vs. WT = -15 ± 4.3% p = 0.04) post-CLP which correlated with significant increase in the level of cardiac oxidative stress following CLP. CONCLUSIONS: In this sepsis model, SE identified cardiomyopathy despite normal EF. SE depression temporally coincides with upregulation of inflammatory cytokines and decreases expression of key mitochondrial ROS scavengers. Upregulation of redox scavenger (CAT) abrogates oxidative stress and cardiac dysfunction in this sepsis model.

9.
Shock ; 47(1): 22-32, 2017 01.
Article in English | MEDLINE | ID: mdl-27488085

ABSTRACT

Necrotizing enterocolitis (NEC) is the most devastating gastrointestinal disease of the premature infant. We have recently shown that NEC development occurs after an increase in proinflammatory CD4Th17 (Th17) cells and reduced anti-inflammatory forkhead box P3 regulatory T cells (Tregs) to the premature small intestine of mice and humans, which can be experimentally reversed in mice by administration of all-trans retinoic acid (ATRA). We have also shown that NEC is characterized by apoptosis of Lgr5-positive intestinal stem cells (ISCs-Lgr5 cells) within the crypts of Lieberkühn, which are subsequently essential for intestinal homeostasis. We now hypothesize that the normal lymphocyte balance within the lamina propria of the intestine can be achieved via administration of ATRA which restores mucosal integrity by preventing the loss of ISCs. Using both in vivo and in vitro strategies, we now demonstrate that Th17 recruitment and Treg depletion lead to increased apoptosis within ISC niches, significantly impairing proliferative capacity and mucosal healing. ATRA exerted its protective effects by preventing T cell imbalance, ultimately leading to the protection of the ISC pool preventing the development of NEC in mice. These findings raise the exciting possibility that dietary manipulations could prevent and treat NEC by modulating lymphocyte balance and the ISC pool within the newborn small intestine.


Subject(s)
Enterocolitis, Necrotizing/drug therapy , Enterocolitis, Necrotizing/prevention & control , Intestines/cytology , Lymphocytes/cytology , Lymphocytes/drug effects , Receptors, G-Protein-Coupled/metabolism , Stem Cells/drug effects , Stem Cells/metabolism , Tretinoin/therapeutic use , Animals , Cell Survival/drug effects , Cell Survival/genetics , Cells, Cultured , Flow Cytometry , Immunohistochemistry , Mice, Inbred C57BL , Real-Time Polymerase Chain Reaction , Stem Cells/cytology
10.
Br J Nutr ; 116(7): 1175-1187, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27609061

ABSTRACT

Necrotising enterocolitis (NEC) is a common disease in premature infants characterised by intestinal ischaemia and necrosis. The only effective preventative strategy against NEC is the administration of breast milk, although the protective mechanisms remain unknown. We hypothesise that an abundant human milk oligosaccharide (HMO) in breast milk, 2'-fucosyllactose (2'FL), protects against NEC by enhancing intestinal mucosal blood flow, and we sought to determine the mechanisms underlying this protection. Administration of HMO-2'FL protected against NEC in neonatal wild-type mice, resulted in a decrease in pro-inflammatory markers and preserved the small intestinal mucosal architecture. These protective effects occurred via restoration of intestinal perfusion through up-regulation of the vasodilatory molecule endothelial nitric oxide synthase (eNOS), as administration of HMO-2'FL to eNOS-deficient mice or to mice that received eNOS inhibitors did not protect against NEC, and by 16S analysis HMO-2'FL affected the microbiota of the neonatal mouse gut, although these changes do not seem to be the primary mechanism of protection. Induction of eNOS by HMO-2'FL was also observed in cultured endothelial cells, providing a link between eNOS and HMO in the endothelium. These data demonstrate that HMO-2'FL protects against NEC in part through maintaining mesenteric perfusion via increased eNOS expression, and suggest that the 2'FL found in human milk may be mediating some of the protective benefits of breast milk in the clinical setting against NEC.


Subject(s)
Enterocolitis, Necrotizing/prevention & control , Infant, Premature, Diseases/physiopathology , Milk, Human/chemistry , Splanchnic Circulation/drug effects , Trisaccharides/administration & dosage , Animals , Animals, Newborn , Disease Models, Animal , Enterocolitis, Necrotizing/physiopathology , Female , Gene Expression/drug effects , Humans , Infant, Newborn , Intestinal Mucosa/blood supply , Mice , Mice, Inbred C57BL , Mice, Knockout , Microbiota/drug effects , Nitric Oxide/analysis , Nitric Oxide Synthase Type III/deficiency , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/physiology
11.
Nat Rev Gastroenterol Hepatol ; 13(10): 590-600, 2016 10.
Article in English | MEDLINE | ID: mdl-27534694

ABSTRACT

Necrotizing enterocolitis (NEC) is the most frequent and lethal disease of the gastrointestinal tract of preterm infants. At present, NEC is thought to develop in the premature host in the setting of bacterial colonization, often after administration of non-breast milk feeds, and disease onset is thought to be due in part to a baseline increased reactivity of the premature intestinal mucosa to microbial ligands as compared with the full-term intestinal mucosa. The increased reactivity leads to mucosal destruction and impaired mesenteric perfusion and partly reflects an increased expression of the bacterial receptor Toll-like receptor 4 (TLR4) in the premature gut, as well as other factors that predispose the intestine to a hyper-reactive state in response to colonizing microorganisms. The increased expression of TLR4 in the premature gut reflects a surprising role for this molecule in the regulation of normal intestinal development through its effects on the Notch signalling pathway. This Review will examine the current approach to the diagnosis and treatment of NEC, provide an overview of our current knowledge regarding its molecular underpinnings and highlight advances made within the past decade towards the development of specific preventive and treatment strategies for this devastating disease.


Subject(s)
Enterocolitis, Necrotizing/prevention & control , Infant, Premature, Diseases/therapy , Animals , Biological Factors/therapeutic use , Biomarkers/metabolism , Breast Feeding , Disease Models, Animal , Disease Susceptibility , Enterocolitis, Necrotizing/diagnosis , Enterocolitis, Necrotizing/etiology , Gastrointestinal Microbiome/physiology , Humans , Infant, Newborn , Infant, Premature , Infant, Premature, Diseases/diagnosis , Infant, Premature, Diseases/etiology , Probiotics/therapeutic use , Treatment Outcome
12.
J Immunol ; 197(3): 859-71, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27307558

ABSTRACT

We seek to define the mechanisms leading to the development of lung disease in the setting of neonatal necrotizing enterocolitis (NEC), a life-threatening gastrointestinal disease of premature infants characterized by the sudden onset of intestinal necrosis. NEC development in mice requires activation of the LPS receptor TLR4 on the intestinal epithelium, through its effects on modulating epithelial injury and repair. Although NEC-associated lung injury is more severe than the lung injury that occurs in premature infants without NEC, the mechanisms leading to its development remain unknown. In this study, we now show that TLR4 expression in the lung gradually increases during postnatal development, and that mice and humans with NEC-associated lung inflammation express higher levels of pulmonary TLR4 than do age-matched controls. NEC in wild-type newborn mice resulted in significant pulmonary injury that was prevented by deletion of TLR4 from the pulmonary epithelium, indicating a role for pulmonary TLR4 in lung injury development. Mechanistically, intestinal epithelial TLR4 activation induced high-mobility group box 1 release from the intestine, which activated pulmonary epithelial TLR4, leading to the induction of the neutrophil recruiting CXCL5 and the influx of proinflammatory neutrophils to the lung. Strikingly, the aerosolized administration of a novel carbohydrate TLR4 inhibitor prevented CXCL5 upregulation and blocked NEC-induced lung injury in mice. These findings illustrate the critical role of pulmonary TLR4 in the development of NEC-associated lung injury, and they suggest that inhibition of this innate immune receptor in the neonatal lung may prevent this devastating complication of NEC.


Subject(s)
Enterocolitis, Necrotizing/complications , Lung Injury/etiology , Respiratory Mucosa/metabolism , Toll-Like Receptor 4/biosynthesis , Animals , Animals, Newborn , Chemokine CXCL5/metabolism , Enterocolitis, Necrotizing/immunology , Enterocolitis, Necrotizing/metabolism , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Gene Knockdown Techniques , HMGB1 Protein/metabolism , Humans , Immunohistochemistry , Infant, Newborn , Lung Injury/immunology , Lung Injury/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Real-Time Polymerase Chain Reaction , Respiratory Mucosa/immunology
13.
Curr Opin Organ Transplant ; 21(2): 178-85, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26867049

ABSTRACT

PURPOSE OF REVIEW: This article discusses the current state of the art in artificial intestine generation in the treatment of short bowel syndrome. RECENT FINDINGS: Short bowel syndrome defines the condition in which patients lack sufficient intestinal length to allow for adequate absorption of nutrition and fluids, and thus need parenteral support. Advances toward the development of an artificial intestine have improved dramatically since the first attempts in the 1980s, and the last decade has seen significant advances in understanding the intestinal stem cell niche, the growth of complex primary intestinal stem cells in culture, and fabrication of the biomaterials that can support the growth and differentiation of these stem cells. There has also been recent progress in understanding the role of the microbiota and the immune cells on the growth of intestinal cultures on scaffolds in animal models. Despite recent progress, there is much work to be done before the development of a functional artificial intestine for short bowel syndrome is successfully achieved. SUMMARY: Continued concerted efforts by cell biologists, bioengineers, and clinician-scientists will be required for the development of an artificial intestine as a clinical treatment modality for short bowel syndrome.


Subject(s)
Intestines/transplantation , Short Bowel Syndrome/surgery , Animals , Cell Culture Techniques , Humans , Intestines/blood supply , Tissue Engineering , Treatment Outcome
14.
Surg Endosc ; 30(9): 3922-7, 2016 09.
Article in English | MEDLINE | ID: mdl-26675939

ABSTRACT

BACKGROUND: Approximately 20-30 % of patients who undergo Roux-en-Y gastric bypass (RYGB) will not meet the goals of weight loss surgery. Revisional surgery is associated with higher morbidity compared to initial operative management, and results in terms of weight loss have been inconsistent. Endoscopic plication has been seen as a less invasive option, with encouraging initial results. The objective was to analyze the outcomes after Restorative Obesity Surgery, Endolumenal (ROSE) procedure. METHODS: We retrospectively analyzed patients who underwent ROSE between 5/2008 and 11/2013. All patients had failure of weight loss or regain weight after RYGB. Demographics, operative data, and follow-up were recorded. RESULTS: Twenty-seven patients underwent ROSE. One patient was excluded due to lack of follow-up. Twenty-five (96 %) patients were female. Mean time since initial RYGB was 11.9 ± 4.3 years. Mean initial weight and BMI were 236 ± 47 lb and 40.6 ± 8.1 kg/m(2), respectively. Mean OR time was 77 ± 30 min. Preoperative average pouch length and stoma diameter were 6.8 ± 2.3 and 2.1 ± 0.7 cm, respectively. On average, 4 ± 1.6 stitches were placed. Final pouch length and stoma diameter were 3.4 ± 1.6 (50 % reduction) and 0.86 ± 0.4 cm (61 % reduction). A total of 12 (46 %) and seven (28 %) patients underwent EGD at 3 and 12 months postoperatively. The mean pouch length and stoma diameter were 5 ± 1.9 (26.5 % reduction) and 1.2 ± 0.7 cm (42.9 % reduction) at 3 months and 6.14 ± 1.6 (10 % reduction) and 2.2 ± 1.2 cm (4.7 % increase) at 12 months, respectively. The %EWL was 8.9, 9.3, 8, 6.7, -10.7, -13.5, -5.8, -4.5 at 3, 6, 12, 24, 36, 48, 60, and 72 months, respectively. CONCLUSION: Although endoscopic plication achieved the intended reduction in the pouch and stoma diameter at 3 months, these tend toward the preoperative diameter at 12 months. This anatomical failure and the lack of follow-up may explain why most patients failed to achieve sustainable weight loss.


Subject(s)
Gastric Bypass/adverse effects , Gastroscopy/methods , Adult , Aged , Female , Humans , Male , Middle Aged , Obesity, Morbid/surgery , Reoperation/methods , Retrospective Studies , Treatment Failure
15.
J Clin Invest ; 126(2): 495-508, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26690704

ABSTRACT

The nature and role of the intestinal leukocytes in necrotizing enterocolitis (NEC), a severe disease affecting premature infants, remain unknown. We now show that the intestine in mouse and human NEC is rich in lymphocytes that are required for NEC development, as recombination activating gene 1­deficient (Rag1­/­) mice were protected from NEC and transfer of intestinal lymphocytes from NEC mice into naive mice induced intestinal inflammation. The intestinal expression of the lipopolysaccharide receptor TLR4, which is higher in the premature compared with full-term human and mouse intestine, is required for lymphocyte influx through TLR4-mediated upregulation of CCR9/CCL25 signaling. TLR4 also mediates a STAT3-dependent polarization toward increased proinflammatory CD3+CD4+IL-17+ and reduced tolerogenic Foxp3+ Treg lymphocytes (Tregs). Th17 lymphocytes were required for NEC development, as inhibition of STAT3 or IL-17 receptor signaling attenuated NEC in mice, while IL-17 release impaired enterocyte tight junctions, increased enterocyte apoptosis, and reduced enterocyte proliferation, leading to NEC. Importantly, TLR4-dependent Th17 polarization could be reversed by the enteral administration of retinoic acid, which induced Tregs and decreased NEC severity. These findings identify an important role for proinflammatory lymphocytes in NEC development via intestinal epithelial TLR4 that could be reversed through dietary modification.


Subject(s)
Enterocolitis, Necrotizing/immunology , Enterocytes/immunology , Infant, Newborn, Diseases/immunology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Toll-Like Receptor 4/immunology , Animals , Enterocolitis, Necrotizing/diet therapy , Enterocolitis, Necrotizing/genetics , Enterocolitis, Necrotizing/pathology , Enterocytes/pathology , Humans , Infant, Newborn , Infant, Newborn, Diseases/diet therapy , Infant, Newborn, Diseases/genetics , Infant, Newborn, Diseases/pathology , Mice , Mice, Knockout , T-Lymphocytes, Regulatory/pathology , Th17 Cells/pathology , Tight Junctions/genetics , Tight Junctions/immunology , Toll-Like Receptor 4/genetics
16.
Innate Immun ; 21(5): 477-89, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25227123

ABSTRACT

Sepsis is a major healthcare problem and a leading cause of death worldwide. There is no dependable diagnosis, and treatment for this condition remains mainly supportive. The etiology of sepsis is related to an overwhelming inflammatory response. In this regard, the antimicrobial protein lipocalin-2 (Lcn2) has been associated with several inflammatory conditions, but its contribution to polymicrobial sepsis is unclear. Polymicrobial sepsis was induced by cecal ligation and puncture (CLP), and Lcn2 mRNA levels and protein expression were measured in liver and lung tissues. We observed that Lcn2 expression was robustly induced in liver and lung of C57BL/6 J (B6) mice, and remained elevated during the stage of innate immune dysfunction observed in sepsis. This response was different in A/J mice, suggesting a contribution of the genetic background, probably due to differences in IL-10 expression between these two mouse strains. Indeed, IL-10 was found to regulate Lcn2 expression in both primary and J774A.1 macrophages. Thus, Lcn2 expression is highly regulated during CLP-induced sepsis, suggesting that this antimicrobial protein could have a role as a potential biomarker for the diagnosis of sepsis.


Subject(s)
Acute-Phase Proteins/biosynthesis , Lipocalins/biosynthesis , Oncogene Proteins/biosynthesis , Sepsis/genetics , Sepsis/microbiology , Acute-Phase Proteins/analysis , Acute-Phase Proteins/genetics , Animals , Biomarkers/analysis , Cecum/injuries , Immunity, Innate/genetics , Interleukin-10/biosynthesis , Interleukin-10/genetics , Ligation , Lipocalin-2 , Lipocalins/analysis , Lipocalins/genetics , Macrophages/metabolism , Mice , Mice, Inbred A , Mice, Inbred C57BL , Oncogene Proteins/analysis , Oncogene Proteins/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Species Specificity
17.
Surg Endosc ; 29(8): 2149-57, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25303921

ABSTRACT

BACKGROUND: Laparoscopic and endoluminal surgical techniques have evolved and allowed improvements in the methods for treating benign and malignant gastrointestinal diseases. To date, only case reports have been reported on the application of a laparo-endoscopic approach for resecting gastric submucosal tumors (SMT). In this study, we aimed to evaluate the efficacy, safety, and oncologic outcomes of a laparo-endoscopic transgastric approach to resect tumors that would traditionally require either a laparoscopic or open surgical approach. Herein, we present the largest single institution series utilizing this technique for the resection of gastric SMT in North America. METHODS: We performed a retrospective review of a prospectively collected patient database. Patients who presented for evaluation of gastric SMT were offered this surgical procedure and informed consents were obtained for participation in the study. RESULTS: Fourteen patients were included in this study between August/2010 and January/2013. Eight (8) patients (57.1 %) were female and the median age was 56 years (range 29-78). Of the 14 cases, 8 patients (57.1 %) underwent laparo-endoscopic resection of SMTs with transgastric extraction, 5 patients (35.7 %) had conversions to traditional laparoscopic surgery, and 1 patient (7.2 %) was abandoned intraoperatively. The median operative time for this cohort was 80 min (range 35-167). Ten patients (71.4 %) had GISTs, 3 (21.4 %) had leiomyomas, and 1 (7.1 %) had schwannoma. There were no intraoperative complications. Two patients had postoperative staple line bleeding that required repeat endoscopy. The median hospital stay was 1 day (range 1-6) and there were no postoperative mortalities. At 12-month follow-up visit, only one GIST patient (10 %) had tumor recurrence. CONCLUSION: Our experience suggests that this surgical approach is safe and efficient in the resection of gastric SMT with transgastric extraction. This study found no intraoperative complications and optimal oncologic outcomes during the follow-up period. Minimally invasive surgical approaches are emerging as a valid and potentially better approach for resecting malignancies; however, continued investigation is underway to further validate this data.


Subject(s)
Gastric Mucosa/surgery , Gastroscopy , Laparoscopy , Stomach Neoplasms/surgery , Adult , Aged , Female , Gastrointestinal Stromal Tumors/surgery , Humans , Leiomyoma/surgery , Length of Stay , Male , Middle Aged , Neoplasm Recurrence, Local , Neurilemmoma/surgery , Operative Time , Retrospective Studies
18.
Shock ; 42(1): 52-9, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24667630

ABSTRACT

Itraconazole (ICZ) is commonly used for the treatment of fungal infections, particularly in immunocompromised patients. In addition, ICZ has been recently found to have antiangiogenic effects and is currently being tested as a new chemotherapeutic agent in several cancer clinical trials. We have previously shown that ICZ impaired complex N-linked glycosylation processing, leading to the accumulation of high-mannose glycoproteins on the surface of macrophages (Møs). This investigation was directed at determining the effects of ICZ on phagocytosis as a major function of Møs. We found a significant decrease in the phagocytosis of opsonized bacterial particles in ICZ-treated murine Møs in comparison with nontreated Møs. Furthermore, the impairment of phagocytosis was associated with a decrease in cell surface expression of Fcγ receptors (FcγRs) as well as alteration of their glycosylation pattern. Concomitantly, a reduction in all three isoforms of the FcγR family (i.e., Fcgr1, Fcgr2, and Fcgr3) mRNA levels was observed after incubation with ICZ. The effect of ICZ on phagocytosis and FcγR expression was reversed by addition of low-density lipoprotein. These studies indicate that ICZ treatment certainly has a dramatic effect on macrophage function, which could result in a potential impairment of the immune system';s ability to respond to pathogens and may lead to an elevated incidence of infections.


Subject(s)
Antifungal Agents/pharmacology , Itraconazole/pharmacology , Phagocytosis/drug effects , Receptors, IgG/antagonists & inhibitors , Animals , Antifungal Agents/administration & dosage , Cell Membrane/metabolism , Cells, Cultured , Cholesterol, LDL/pharmacology , Dose-Response Relationship, Drug , Escherichia coli/immunology , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Glycosylation/drug effects , Itraconazole/administration & dosage , Itraconazole/antagonists & inhibitors , Macrophages/drug effects , Macrophages/immunology , Male , Mice, Inbred Strains , Receptors, IgG/genetics , Receptors, IgG/metabolism
19.
Innate Immun ; 19(1): 30-41, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22751446

ABSTRACT

Scavenger receptor A (Sra), also known as macrophage scavenger receptor 1 (Msr1), is a surface glycoprotein preferentially present in macrophages that plays a primary role in innate immunity. Previous studies have shown that Sra is a modifier gene for the response to bacterial LPS in mice at the level of IL-10 production, in particular. In the present study, we found that Sra(-/-) mice are more resistant to septic shock induced by cecal ligation and puncture than wild-type C57BL/6 J (B6) mice. In addition, Sra(-/-) mice displayed initial elevated high density lipoprotein (HDL) circulating levels. Naïve peritoneal macrophages (PMs) were isolated from Sra(-/-) mice to understand the possible protective mechanism. Incubation of these cells with LPS was found to modulate TLR4 signaling, leading to a reduction in IL-10 and IL-6 mRNA levels, but not TNF-α expression, at low concentrations of LPS in comparison with PMs isolated from B6 mice. No differences were found in LPS binding between PMs derived from Sra(-/-) or B6 mice. The lack of Sra binding to LPS was confirmed after transfection of Chinese hamster ovary (CHO) cells with the Sra gene. The contribution of Sra to the outcome of sepsis may be a combination of changes in TLR4 signaling pathway and elevated levels of HDL in circulation, but also LPS toxicity.


Subject(s)
Cytokines/metabolism , Macrophages, Peritoneal/immunology , Scavenger Receptors, Class A/metabolism , Shock, Septic/immunology , Toll-Like Receptor 4/metabolism , Animals , CHO Cells , Cricetinae , Lipopolysaccharides/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Scavenger Receptors, Class A/genetics , Shock, Septic/genetics , Signal Transduction/genetics , Signal Transduction/immunology
20.
J Surg Res ; 174(2): 344-51, 2012 May 15.
Article in English | MEDLINE | ID: mdl-21474146

ABSTRACT

BACKGROUND: Sepsis is a major health problem in the United States that affects more than three-quarters of a million people every year. Previous studies have shown that scavenger receptor A (Sra), also known as macrophage scavenger receptor 1 (Msr1), is a modifier of interleukin 10 (IL-10) expression after injection of bacterial lipopolysaccharide (LPS). Therefore, we investigated the response to sepsis in Sra knock out mice. MATERIALS AND METHODS: C57BL/6J (B6) (n = 88) and Sra (-/-) mice (n = 88) were subjected to cecal ligation and puncture (CLP) using 18G or 16G needles, sham operation, or non-operated controls. At the end, mice were autopsied for the determination of abnormalities after the procedure. Cytokine gene expression was examined in lung and liver samples by quantitative RT-PCR (qRT-PCR), and circulating cholesterol levels were also measured. RESULTS: Sra (-/-) mice displayed an enlargement of the gallbladder after CLP that was not detected in sham or non-operated mice or in B6 mice (wild-type) after CLP. The enlarged gallbladder resembles a condition of acute acalculous cholecystitis observed in humans. Sra (-/-) mice presented high cholesterol levels in circulation as opposed to wild type B6 mice. Moreover, Sra (-/-) mice exhibited a reduction in IL-10 mRNA levels in lungs compared to wild-type B6 mice after CLP. CONCLUSIONS: The development of acute acalculous cholecystitis may be the combination of pre-existing conditions, such as hypercholesterolemia associated with a defect in Sra (Msr1) and a robust inflammation induced by sepsis.


Subject(s)
Acalculous Cholecystitis/etiology , Scavenger Receptors, Class A/genetics , Sepsis/complications , Acalculous Cholecystitis/metabolism , Animals , Cecum/surgery , Cholesterol/blood , Disease Models, Animal , Interleukin-10/metabolism , Ligation , Lung/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Sepsis/genetics , Sepsis/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...