101
|
McCleary EJ, Tajchman S. Parenteral Nutrition and Infection Risk in the Intensive Care Unit: A Practical Guide for the Bedside Clinician. Nutr Clin Pract 2016; 31:476-89. [PMID: 27317614 DOI: 10.1177/0884533616653808] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The safety of parenteral nutrition (PN) administration in critically ill patients has been the subject of much controversy. Historically, PN administration has been associated with an increased risk of bacterial and fungal infections, leading to significant morbidity and mortality. Much of the data showing increased infectious complications compared with either no nutrition or enteral nutrition was derived from early studies conducted in the 1980s-2000s. Poor glucose control and hyperalimentation are confounding factors in many early studies, making it difficult to determine the true PN infection risks. While PN studies conducted during the past 10 years have failed to show the same infection rates, these risks continue to be cited as dogma. Potential reasons for such discordant results include improved glycemic control, avoidance of overfeeding, and improved sterility and central venous catheter care. Understanding the true infectious risk of PN administration in the intensive care unit is necessary to optimize patient care, as inappropriately withholding such nutrition is potentially deleterious. This review is meant to serve as a practical guide to the bedside clinician who is evaluating the risks and benefits of initiating PN in a critically ill patient. Each component of PN will be evaluated based on risk of infection, and the potential ways to mitigate risks will be discussed.
Collapse
Affiliation(s)
- Emily J McCleary
- University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Sharla Tajchman
- University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
102
|
Rugeles S, Villarraga-Angulo LG, Ariza-Gutiérrez A, Chaverra-Kornerup S, Lasalvia P, Rosselli D. High-protein hypocaloric vs normocaloric enteral nutrition in critically ill patients: A randomized clinical trial. J Crit Care 2016; 35:110-4. [PMID: 27481744 DOI: 10.1016/j.jcrc.2016.05.004] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 04/29/2016] [Accepted: 05/02/2016] [Indexed: 01/04/2023]
Abstract
PURPOSE Appropriate caloric intake in critically ill patients receiving enteral nutrition is controversial. This study evaluates the impact of different caloric regimens on severity of organ failure measured with Sequential Organ Failure Assessment (SOFA). MATERIALS AND METHODS We conducted a randomized prospective controlled trial. Study population included adult intensive care unit (ICU) patients expected to require enteral nutrition for more than 96 hours. Goals in the intervention group were hypocaloric (15 kcal/kg per day) enteral nutrition compared to normocaloric (25 kcal/kg per day) enteral nutrition, both with hyperproteic intake (1.7 g of protein/kg per day). Primary end point was change in SOFA score (ΔSOFA) from baseline at 48 hours. Secondary end points were ΔSOFA at 96 hours, insulin requirements, hyperglycemia or hypoglycemic episodes, length of ICU stay, days on ventilator, and 28-day mortality. RESULTS After screening 443 patients, 120 patients were analyzed. There were no differences between groups in baseline characteristics. We did not find a statistically significant difference in ΔSOFA at 48 hours. Patients in the hypocaloric group showed lower average daily insulin requirements and percentage of patients requiring any insulin. CONCLUSIONS Hyperproteic, hypocaloric nutrition did not show different outcomes compared to normocaloric nutrition, except lower insulin requirements. Hypocaloric nutrition could provide a more physiologic approach with lower need for care and metabolic impact.
Collapse
Affiliation(s)
- Saúl Rugeles
- Surgery Department, Pontificia Universidad Javeriana, Medical School, Hospital Universitario San Ignacio, Bogota, Colombia.
| | - Luis Gabriel Villarraga-Angulo
- Surgery Department, Pontificia Universidad Javeriana, Medical School, Hospital Universitario San Ignacio, Bogota, Colombia.
| | - Aníbal Ariza-Gutiérrez
- Surgery Department, Pontificia Universidad Javeriana, Medical School, Hospital Universitario San Ignacio, Bogota, Colombia.
| | | | | | - Diego Rosselli
- Clinical Epidemiology and Biostatistics Department, Pontificia Universidad Javeriana, Medical School, Bogota, Colombia.
| |
Collapse
|
103
|
Abstract
Intestinal failure (IF) is a state in which the nutritional demands are not met by the gastrointestinal absorptive surface. A majority of IF cases are associated with short-bowel syndrome, which is a result of malabsorption after significant intestinal resection for numerous reasons, some of which include Crohn's disease, vascular thrombosis, and radiation enteritis. IF can also be caused by obstruction, dysmotility, and congenital defects. Recognition and management of IF can be challenging, given the complex nature of this condition. This review discusses the management of IF with a focus on intestinal rehabilitation, parenteral nutrition, and transplantation.
Collapse
|
104
|
|
105
|
Elke G, van Zanten ARH, Lemieux M, McCall M, Jeejeebhoy KN, Kott M, Jiang X, Day AG, Heyland DK. Enteral versus parenteral nutrition in critically ill patients: an updated systematic review and meta-analysis of randomized controlled trials. Crit Care 2016; 20:117. [PMID: 27129307 PMCID: PMC4851818 DOI: 10.1186/s13054-016-1298-1] [Citation(s) in RCA: 222] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 04/14/2016] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Enteral nutrition (EN) is recommended as the preferred route for early nutrition therapy in critically ill adults over parenteral nutrition (PN). A recent large randomized controlled trial (RCT) showed no outcome differences between the two routes. The objective of this systematic review was to evaluate the effect of the route of nutrition (EN versus PN) on clinical outcomes of critically ill patients. METHODS An electronic search from 1980 to 2016 was performed identifying relevant RCTs. Individual trial data were abstracted and methodological quality of included trials scored independently by two reviewers. The primary outcome was overall mortality and secondary outcomes included infectious complications, length of stay (LOS) and mechanical ventilation. Subgroup analyses were performed to examine the treatment effect by dissimilar caloric intakes, year of publication and trial methodology. We performed a test of asymmetry to assess for the presence of publication bias. RESULTS A total of 18 RCTs studying 3347 patients met inclusion criteria. Median methodological score was 7 (range, 2-12). No effect on overall mortality was found (1.04, 95 % CI 0.82, 1.33, P = 0.75, heterogeneity I(2) = 11 %). EN compared to PN was associated with a significant reduction in infectious complications (RR 0.64, 95 % CI 0.48, 0.87, P = 0.004, I(2) = 47 %). This was more pronounced in the subgroup of RCTs where the PN group received significantly more calories (RR 0.55, 95 % CI 0.37, 0.82, P = 0.003, I(2) = 0 %), while no effect was seen in trials where EN and PN groups had a similar caloric intake (RR 0.94, 95 % CI 0.80, 1.10, P = 0.44, I(2) = 0 %; test for subgroup differences, P = 0.003). Year of publication and methodological quality did not influence these findings; however, a publication bias may be present as the test of asymmetry was significant (P = 0.003). EN was associated with significant reduction in ICU LOS (weighted mean difference [WMD] -0.80, 95 % CI -1.23, -0.37, P = 0.0003, I(2) = 0 %) while no significant differences in hospital LOS and mechanical ventilation were observed. CONCLUSIONS In critically ill patients, the use of EN as compared to PN has no effect on overall mortality but decreases infectious complications and ICU LOS. This may be explained by the benefit of reduced macronutrient intake rather than the enteral route itself.
Collapse
Affiliation(s)
- Gunnar Elke
- Department of Anaesthesiology and Intensive Care Medicine, University Medical Centre Schleswig-Holstein, Campus Kiel, Arnold-Heller-Str. 3 Haus 12, 24105, Kiel, Germany
| | - Arthur R H van Zanten
- Department of Intensive Care, Gelderse Vallei Hospital, Willy Brandtlaan 10, 6716, RP, Ede, The Netherlands
| | - Margot Lemieux
- Department of Critical Care Medicine, Queen's University and Clinical Evaluation Research Unit, Kingston General Hospital, Angada 4, K7L 2V7, Kingston, ON, Canada
| | - Michele McCall
- Medical/Surgical ICU, Specialized Complex Care, St Michael's Hospital, 30 Bond Street, Toronto, ON, Canada
| | - Khursheed N Jeejeebhoy
- Department of Nutritional Sciences, St Michael's Hospital, 30 Bond Street, Toronto, ON, Canada
| | - Matthias Kott
- Department of Anaesthesiology and Intensive Care Medicine, University Medical Centre Schleswig-Holstein, Campus Kiel, Arnold-Heller-Str. 3 Haus 12, 24105, Kiel, Germany
| | - Xuran Jiang
- Department of Critical Care Medicine, Queen's University and Clinical Evaluation Research Unit, Kingston General Hospital, Angada 4, K7L 2V7, Kingston, ON, Canada
| | - Andrew G Day
- Department of Critical Care Medicine, Queen's University and Clinical Evaluation Research Unit, Kingston General Hospital, Angada 4, K7L 2V7, Kingston, ON, Canada
| | - Daren K Heyland
- Department of Critical Care Medicine, Queen's University and Clinical Evaluation Research Unit, Kingston General Hospital, Angada 4, K7L 2V7, Kingston, ON, Canada.
| |
Collapse
|
106
|
Evans DC, Forbes R, Jones C, Cotterman R, Njoku C, Thongrong C, Tulman D, Bergese SD, Thomas S, Papadimos TJ, Stawicki SP. Continuous versus bolus tube feeds: Does the modality affect glycemic variability, tube feeding volume, caloric intake, or insulin utilization? Int J Crit Illn Inj Sci 2016; 6:9-15. [PMID: 27051616 PMCID: PMC4795366 DOI: 10.4103/2229-5151.177357] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Introduction: Enteral nutrition (EN) is very important to optimizing outcomes in critical illness. Debate exists regarding the best strategy for enteral tube feeding (TF), with concerns that bolus TF (BTF) may increase glycemic variability (GV) but result in fewer nutritional interruptions than continuous TF (CTF). This study examines if there is a difference in GV, insulin usage, TF volume, and caloric delivery among intensive care patients receiving BTF versus CTF. We hypothesize that there are no significant differences between CTF and BTF when comparing the above parameters. Materials and Methods: Prospective, randomized pilot study of critically ill adult patients undergoing percutaneous endoscopic gastrostomy (PEG) placement for EN was performed between March 1, 2012 and May 15, 2014. Patients were randomized to BTF or CTF. Glucose values, insulin use, TF volume, and calories administered were recorded. Data were organized into 12-h epochs for statistical analyses and GV determination. In addition, time to ≥80% nutritional delivery goal, demographics, Acute Physiology and Chronic Health Evaluation II scores, and TF interruptions were examined. When performing BTF versus CTF assessments, continuous parameters were compared using Mann–Whitney U-test or repeated measures t-test, as appropriate. Categorical data were analyzed using Fisher's exact test. Results: No significant demographic or physiologic differences between the CTF (n = 24) and BTF (n = 26) groups were seen. The immediate post-PEG 12-h epoch showed significantly lower GV and median TF volume for patients in the CTF group. All subsequent epochs (up to 18 days post-PEG) showed no differences in GV, insulin use, TF volume, or caloric intake. Insulin use for both groups increased when comparing the first 24 h post-PEG values to measurements from day 8. There were no differences in TF interruptions, time to ≥80% nutritional delivery goal, or hypoglycemic episodes. Conclusions: This study demonstrated no clinically relevant differences in GV, insulin use, TF volume or caloric intake between BTF and CTF groups. Despite some shortcomings, our data suggest that providers should not feel limited to BTF or CTF because of concerns for GV, time to goal nutrition, insulin use, or caloric intake, and should consider other factors such as resource utilization, ease of administration, and/or institutional/patient characteristics.
Collapse
Affiliation(s)
- David C Evans
- Department of Surgery, Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Rachel Forbes
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Christian Jones
- Department of Surgery, Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Robert Cotterman
- Department of Surgery, Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Chinedu Njoku
- Department of Surgery, Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Cattleya Thongrong
- Department of Anesthesiology, Ohio State University College of Medicine, Columbus, Ohio, USA
| | - David Tulman
- Department of Anesthesiology, Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Sergio D Bergese
- Department of Anesthesiology, Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Sheela Thomas
- Department of Clinical Nutrition, Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Thomas J Papadimos
- Department of Anesthesiology, Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Stanislaw P Stawicki
- Department of Research and Innovation, St. Luke's University Hospital, Bethlehem, Pennsylvania, USA
| |
Collapse
|
107
|
|
108
|
Wirth R, Dziewas R, Beck AM, Clavé P, Hamdy S, Heppner HJ, Langmore S, Leischker AH, Martino R, Pluschinski P, Rösler A, Shaker R, Warnecke T, Sieber CC, Volkert D. Oropharyngeal dysphagia in older persons - from pathophysiology to adequate intervention: a review and summary of an international expert meeting. Clin Interv Aging 2016; 11:189-208. [PMID: 26966356 PMCID: PMC4770066 DOI: 10.2147/cia.s97481] [Citation(s) in RCA: 308] [Impact Index Per Article: 34.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Oropharyngeal dysphagia (OD) is a highly prevalent and growing condition in the older population. Although OD may cause very severe complications, it is often not detected, explored, and treated. Older patients are frequently unaware of their swallowing dysfunction which is one of the reasons why the consequences of OD, ie, aspiration, dehydration, and malnutrition, are regularly not attributed to dysphagia. Older patients are particularly vulnerable to dysphagia because multiple age-related changes increase the risk of dysphagia. Physicians in charge of older patients should be aware that malnutrition, dehydration, and pneumonia are frequently caused by (unrecognized) dysphagia. The diagnosis is particularly difficult in the case of silent aspiration. In addition to numerous screening tools, videofluoroscopy was the traditional gold standard of diagnosing OD. Recently, the fiberoptic endoscopic evaluation of swallowing is increasingly utilized because it has several advantages. Besides making a diagnosis, fiberoptic endoscopic evaluation of swallowing is applied to evaluate the effectiveness of therapeutic maneuvers and texture modification of food and liquids. In addition to swallowing training and nutritional interventions, newer rehabilitation approaches of stimulation techniques are showing promise and may significantly impact future treatment strategies.
Collapse
Affiliation(s)
- Rainer Wirth
- Department for Internal Medicine and Geriatrics, St Marien-Hospital Borken, Borken, Germany; Institute for Biomedicine of Aging, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nürnberg, Germany
| | - Rainer Dziewas
- Department of Neurology, University Hospital Münster, Münster, Germany
| | - Anne Marie Beck
- Department of Nutrition and Health, Faculty of Health and Technology, Metropolitan University College, Copenhagen, Denmark
| | - Pere Clavé
- Centro de Investigación Biomédica en Red de enfermadades Hepáticas y Digestivas (CIBERehd), Hospital de Mataró, Universitat Autònoma de Barcelona, Mataró, Spain
| | - Shaheen Hamdy
- Centre for Gastrointestinal Sciences, Institute of Inflammation and Repair, University of Manchester, Salford Royal Hospital, Salford, UK
| | - Hans Juergen Heppner
- Department of Geriatrics, Witten- Herdecke University, Schwelm, Germany; Helios Clinic Schwelm, Schwelm, Germany
| | - Susan Langmore
- Department of Speech, Language and Hearing Sciences, Boston University School of Medicine, Boston, MA, USA
| | | | - Rosemary Martino
- Department of Speech-Language Pathology, University of Toronto, Toronto, Canada
| | - Petra Pluschinski
- Department of Phoniatrics and Pediatric Audiology, University of Marburg, Marburg, Germany
| | - Alexander Rösler
- Department of Geriatrics, Marien Hospital Hamburg, Hamburg, Germany
| | - Reza Shaker
- Division of Gastroenterology and Hepatology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Tobias Warnecke
- Department of Neurology, University Hospital Münster, Münster, Germany
| | - Cornel Christian Sieber
- Institute for Biomedicine of Aging, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nürnberg, Germany; Department of General Internal Medicine and Geriatrics, St John of God Hospital Regensburg, Regensburg, Germany
| | - Dorothee Volkert
- Institute for Biomedicine of Aging, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nürnberg, Germany
| |
Collapse
|
109
|
Feng Y, Barrett M, Hou Y, Yoon HK, Ochi T, Teitelbaum DH. Homeostasis alteration within small intestinal mucosa after acute enteral refeeding in total parenteral nutrition mouse model. Am J Physiol Gastrointest Liver Physiol 2016; 310:G273-84. [PMID: 26635320 PMCID: PMC4754738 DOI: 10.1152/ajpgi.00335.2015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 11/24/2015] [Indexed: 01/31/2023]
Abstract
Feeding strategies to care for patients who transition from enteral nutrient deprivation while on total parenteral nutrition (TPN) to enteral feedings generally proceed to full enteral nutrition once the gastrointestinal tract recovers; however, an increasing body of literature suggests that a subgroup of patients may actually develop an increased incidence of adverse events, including death. To examine this further, we studied the effects of acute refeeding in a mouse model of TPN. Interestingly, refeeding led to some beneficial effects, including prevention in the decline in intestinal epithelial cell (IEC) proliferation. However, refeeding led to a significant increase in mucosal expression of proinflammatory cytokines, including tumor necrosis factor-α (TNF-α), as well as an upregulation in Toll-like receptor 4 (TLR-4). Refeeding also failed to prevent TPN-associated increases in IEC apoptosis, loss of epithelial barrier function, and failure of the leucine-rich repeat-containing G protein-coupled receptor 5-positive stem cell expression. Transitioning from TPN to enteral feedings led to a partial restoration of the small bowel microbial population. In conclusion, while acute refeeding led to some restoration of normal gastrointestinal physiology, enteral refeeding led to a significant increase in mucosal inflammatory markers and may suggest alternative strategies to enteral refeeding should be considered.
Collapse
Affiliation(s)
- Yongjia Feng
- 1Section of Pediatric Surgery, Department of Surgery, the University of Michigan Medical School and the C. S. Mott Children's Hospital, Ann Arbor, Michigan;
| | - Meredith Barrett
- 1Section of Pediatric Surgery, Department of Surgery, the University of Michigan Medical School and the C. S. Mott Children's Hospital, Ann Arbor, Michigan; ,2General Surgery, Department of Surgery, the University of Michigan Medical School, Ann Arbor, Michigan;
| | - Yue Hou
- 1Section of Pediatric Surgery, Department of Surgery, the University of Michigan Medical School and the C. S. Mott Children's Hospital, Ann Arbor, Michigan; ,3University of Michigan, Ann Arbor, Michigan; and
| | - Hong Keun Yoon
- 1Section of Pediatric Surgery, Department of Surgery, the University of Michigan Medical School and the C. S. Mott Children's Hospital, Ann Arbor, Michigan; ,3University of Michigan, Ann Arbor, Michigan; and
| | - Takanori Ochi
- 1Section of Pediatric Surgery, Department of Surgery, the University of Michigan Medical School and the C. S. Mott Children's Hospital, Ann Arbor, Michigan; ,4Department of Pediatric Surgery, Juntendo Hospital, Juntendo University, Tokyo, Japan
| | - Daniel H. Teitelbaum
- 1Section of Pediatric Surgery, Department of Surgery, the University of Michigan Medical School and the C. S. Mott Children's Hospital, Ann Arbor, Michigan;
| |
Collapse
|
110
|
Classification, prevention and management of entero-atmospheric fistula: a state-of-the-art review. Langenbecks Arch Surg 2016; 401:1-13. [PMID: 26867939 DOI: 10.1007/s00423-015-1370-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 12/22/2015] [Indexed: 02/07/2023]
Abstract
BACKGROUND Entero-atmospheric fistula (EAF) is an enteric fistula occurring in the setting of an open abdomen, thus creating a communication between the GI tract and the external atmosphere. Management and nursing of patients suffering EAF carries several challenges, and prevention of EAF should be the first and best treatment option. PURPOSE Here, we present a novel modified classification of EAF and review the current state of the art in its prevention and management including nutritional issues and feeding strategies. We also provide an overview on surgical management principles, highlighting several surgical techniques for dealing with EAF that have been reported in the literature throughout the years. CONCLUSIONS The treatment strategy for EAF should be multidisciplinary and multifaceted. Surgical treatment is most often multistep and should be tailored to the single patient, based on the type and characteristics of the EAF, following its correct identification and classification. The specific experience of surgeons and nursing staff in the management of EAF could be enhanced, applying distinct simulation-based ex vivo training models.
Collapse
|
111
|
McClave SA, Taylor BE, Martindale RG, Warren MM, Johnson DR, Braunschweig C, McCarthy MS, Davanos E, Rice TW, Cresci GA, Gervasio JM, Sacks GS, Roberts PR, Compher C. Guidelines for the Provision and Assessment of Nutrition Support Therapy in the Adult Critically Ill Patient: Society of Critical Care Medicine (SCCM) and American Society for Parenteral and Enteral Nutrition (A.S.P.E.N.). JPEN J Parenter Enteral Nutr 2016; 40:159-211. [PMID: 26773077 DOI: 10.1177/0148607115621863] [Citation(s) in RCA: 1835] [Impact Index Per Article: 203.9] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Stephen A McClave
- Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Beth E Taylor
- Nutrition Support Specialist, Barnes Jewish Hospital, St Louis, Missouri
| | - Robert G Martindale
- Chief Division of General Surgery, Oregon Health and Science University, Portland, Oregon
| | - Malissa M Warren
- Critical Care Dietitian, Portland VA Medical Center, Portland, Oregon
| | - Debbie R Johnson
- Clinical Nurse Specialist: Wound, Skin, Ostomy, UW Health University of Wisconsin Hospital and Clinics, Madison, Wisconsin
| | - Carol Braunschweig
- Professor, Department of Kinesiology and Nutrition and Division of Epidemiology and Biostatistics, University of Illinois at Chicago, Chicago, Illinois
| | - Mary S McCarthy
- Senior Nurse Scientist, Center for Nursing Science and Clinical Inquiry, Madigan Healthcare System, Tacoma, Washington
| | - Evangelia Davanos
- Pharmacotherapy Specialist, Nutrition Support, The Brooklyn Hospital Center, Brooklyn, New York
| | - Todd W Rice
- Assistant Professor of Medicine, Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Gail A Cresci
- Project Research Staff, Digestive Disease Institute, Gastroenterology and Pathobiology, Cleveland, Ohio
| | - Jane M Gervasio
- Chair and Professor of Pharmacy Practice, Butler University College of Pharmacy and Health Science, Indianapolis, Indiana
| | - Gordon S Sacks
- Professor and Head, Department of Pharmacy Practice, Harrison School of Pharmacy, Auburn University, Auburn, Alabama
| | - Pamela R Roberts
- Professor and Vice Chair, Division Chief of Critical Care Medicine, Director of Research John A. Moffitt Endowed Chair, Department of Anesthesiology, Oklahoma City, Oklahoma
| | - Charlene Compher
- Professor of Nutrition Science, University of Pennsylvania School of Nursing, Philadelphia, Pennsylvania
| | | | | |
Collapse
|
112
|
Epidemiological and clinical features for cefepime heteroresistant Escherichia coli infections in Southwest China. Eur J Clin Microbiol Infect Dis 2016; 35:571-8. [PMID: 26815433 DOI: 10.1007/s10096-015-2572-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 12/30/2015] [Indexed: 01/13/2023]
Abstract
Phenotypic heteroresistance (PHR) is common in a weight of microbes and plays an important role in the evolution of antibiotic resistance. However, PHR to cefepime (FEP-PHR) among invasive Escherichia coli (E. coli) has not been reported. This study aimed to report the characteristics of invasive E. coli with FEP-PHR traits and further to investigate the predisposing factors for its acquisition. A retrospective case-control study was conducted in a teaching hospital in Southwest China. A total of 319 successive and non-duplicate E. coli strains were isolated from blood and other sterile body fluids between July 2011 and August 2013. Among the seventy (70/319, 21.9 %) isolates harboring FEP-PHR traits, 30 (42.9 %) isolates were isolated from blood, 14 (20.0 %) isolates were isolated from bile, and 13 (18.6 %) isolates were isolated from drainage. FEP-PHR isolates were verified by population analysis profile (PAP) assays. Male gender, receipt of total parenteral nutrition, cephalosporins exposure, and production of extended spectrum betalactamases (ESBL) were independent risk factors for the acquisition of invasive E. coli with FEP-PHR traits. Pulsedfield gel electrophoresis (PFGE) revealed clonal diversity among the FEP-PHR isolates. The prevalence of heteroresistance to cefepime among invasive E. coli isolates merits great attention and heteroresistance may lead to the emergence of resistance strains. Therefore, systematical analysis of risk factors, careful interpretation of antibiotic susceptibility results and appropriate prescription of therapeutic strategy could help to prevent misreporting and therapeutic failure.
Collapse
|
113
|
GLP-2 Prevents Intestinal Mucosal Atrophy and Improves Tissue Antioxidant Capacity in a Mouse Model of Total Parenteral Nutrition. Nutrients 2016; 8:nu8010033. [PMID: 26761030 PMCID: PMC4728647 DOI: 10.3390/nu8010033] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 01/03/2016] [Accepted: 01/05/2016] [Indexed: 01/04/2023] Open
Abstract
We investigated the effects of exogenous glucagon-like peptide-2 (GLP-2) on mucosal atrophy and intestinal antioxidant capacity in a mouse model of total parenteral nutrition (TPN). Male mice (6–8 weeks old) were divided into three groups (n = 8 for each group): a control group fed a standard laboratory chow diet, and experimental TPN (received standard TPN solution) and TPN + GLP-2 groups (received TPN supplemented with 60 µg/day of GLP-2 for 5 days). Mice in the TPN group had lower body weight and reduced intestinal length, villus height, and crypt depth compared to the control group (all p < 0.05). GLP-2 supplementation increased all parameters compared to TPN only (all p < 0.05). Intestinal total superoxide dismutase activity and reduced-glutathione level in the TPN + GLP-2 group were also higher relative to the TPN group (all p < 0.05). GLP-2 administration significantly upregulated proliferating cell nuclear antigen expression and increased glucose-regulated protein (GRP78) abundance. Compared with the control and TPN + GLP-2 groups, intestinal cleaved caspase-3 was increased in the TPN group (all p < 0.05). This study shows GLP-2 reduces TPN-associated intestinal atrophy and improves tissue antioxidant capacity. This effect may be dependent on enhanced epithelial cell proliferation, reduced apoptosis, and upregulated GRP78 expression.
Collapse
|
114
|
Japanese Guidelines for Nutrition Support Therapy in the Adult and Pediatric Critically Ill Patients. ACTA ACUST UNITED AC 2016. [DOI: 10.3918/jsicm.23.185] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
115
|
Abstract
Early provision of enteral nutrition (EN) in critically ill and injured patients has become standard practice in surgical intensive care units (ICUs) due to its proven role in reducing septic complications. Increasingly, intensivists are confronted with patients with an open abdomen due to the use of damage control surgery and the recognition of the abdominal compartment syndrome; the role and timing of EN in these challenging patients continue to be debated. Patients with an open abdomen are often among the sickest in the ICU and hence could benefit from early nutrition support. However, the exposed abdominal viscera can understandably create anxiety regarding the initiation of EN; there is theoretic concern over exacerbation of bowel distention with resultant inability to close the abdomen and an increased aspiration risk due to paralytic ileus. Recent studies have investigated the utility of EN in the patient with an open abdomen, addressing these clinical concerns. The goal of this clinical review is to provide guidance to physicians caring for these complex patients.
Collapse
Affiliation(s)
- Scott M Moore
- Department of Surgery, Denver Health Medical Center, University of Colorado, Denver, Colorado, USA
| | - Clay Cothren Burlew
- Department of Surgery, Denver Health Medical Center, University of Colorado, Denver, Colorado, USA
| |
Collapse
|
116
|
Malik AA, Rajandram R, Tah PC, Hakumat-Rai VR, Chin KF. Microbial cell preparation in enteral feeding in critically ill patients: A randomized, double-blind, placebo-controlled clinical trial. J Crit Care 2015; 32:182-8. [PMID: 26777745 DOI: 10.1016/j.jcrc.2015.12.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 11/05/2015] [Accepted: 12/08/2015] [Indexed: 12/24/2022]
Abstract
Gut failure is a common condition in critically ill patients in the intensive care unit (ICU). Enteral feeding is usually the first line of choice for nutrition support in critically ill patients. However, enteral feeding has its own set of complications such as alterations in gut transit time and composition of gut eco-culture. The primary aim of this study was to investigate the effect of microbial cell preparation on the return of gut function, white blood cell count, C-reactive protein levels, number of days on mechanical ventilation, and length of stay in ICU. A consecutive cohort of 60 patients admitted to the ICU in University Malaya Medical Centre requiring enteral feeding were prospectively randomized to receive either treatment (n = 30) or placebo (n = 30). Patients receiving enteral feeding supplemented with a course of treatment achieved a faster return of gut function and required shorter duration of mechanical ventilation and shorter length of stay in the ICU. However, inflammatory markers did not show any significant change in the pretreatment and posttreatment groups. Overall, it can be concluded that microbial cell preparation enhances gut function and the overall clinical outcome of critically ill patients receiving enteral feeding in the ICU.
Collapse
Affiliation(s)
- Ausama A Malik
- Department of Surgery, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Retnagowri Rajandram
- Department of Surgery, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Pei Chien Tah
- Department of Dietetics, University of Malaya Medical Centre, Kuala Lumpur, Malaysia
| | - Vineya-Rai Hakumat-Rai
- Department of Anesthesiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Kin-Fah Chin
- Department of Surgery, Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Selangor, Malaysia.
| |
Collapse
|
117
|
Puiggròs C, Molinos R, Ortiz MD, Ribas M, Romero C, Vázquez C, Segurola H, Burgos R. Experience in Bedside Placement, Clinical Validity, and Cost-Efficacy of a Self-Propelled Nasojejunal Feeding Tube. Nutr Clin Pract 2015; 30:815-23. [PMID: 26214512 PMCID: PMC4708005 DOI: 10.1177/0884533615592954] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND The procedures needed to insert nasojejunal tubes (NJTs) are often invasive or uncomfortable for the patient and require hospital resources. The objectives of this study were to describe our experience in inserting a self-propelling NJT with distal pigtail end and evaluate clinical validity and cost efficacy of this enteral nutrition (EN) approach compared with parenteral nutrition (PN). MATERIALS AND METHODS Prospective study from July 2009 to December 2010, including hospitalized noncritical patients who required short-term jejunal EN. The tubes were inserted at bedside, using intravenous erythromycin as a prokinetic drug. Positioning was considered correct when the distal end was beyond the ligament of Treitz. Migration failure was considered when the tube was not positioned into the jejunum within 48 hours postinsertion. RESULTS Fifty-six insertions were recorded in 47 patients, most frequently in severe acute pancreatitis (69.6%). The migration rates at 18 and 48 hours postinsertion were 73.2% and 82.1%, respectively. There was migration failure in 8.9% of cases, and 8.9% were classified null (the tube was no longer in the gastrointestinal tract at 18 hours). There were no reported or observed complications. The mean duration of the EN was 12 ± 10.8 days. Five different types of EN formula were used. The total study cost was 53.9% lower compared with using PN in all patients. CONCLUSIONS Our study demonstrated that bedside insertion of a self-propelling NJT is a safe, cost-effective, and successful technique for postpyloric enteral feeding in at least 73% of the patients, and only 18% of patients could eventually need other placement techniques. It can avoid the need for more aggressive or expensive placement techniques or even PN if we cannot achieve enteral access.
Collapse
Affiliation(s)
- Carolina Puiggròs
- Nutritional Support Unit, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Rosa Molinos
- Nutritional Support Unit, Vall d'Hebron University Hospital, Barcelona, Spain
| | - M Dolors Ortiz
- Nutritional Support Unit, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Montserrat Ribas
- Nutritional Support Unit, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Carlos Romero
- Nutritional Support Unit, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Concepcion Vázquez
- Nutritional Support Unit, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Hegoi Segurola
- Nutritional Support Unit, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Rosa Burgos
- Nutritional Support Unit, Vall d'Hebron University Hospital, Barcelona, Spain
| |
Collapse
|
118
|
Wang D, Zheng SQ, Chen XC, Jiang SW, Chen HB. Comparisons between small intestinal and gastric feeding in severe traumatic brain injury: a systematic review and meta-analysis of randomized controlled trials. J Neurosurg 2015; 123:1194-201. [PMID: 26024007 DOI: 10.3171/2014.11.jns141109] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
OBJECT Nutritional support is highly recommended for reducing the risk of nosocomial infections, such as pneumonitis, in patients with severe traumatic brain injury (TBI). Currently, there is no consensus for the preferred route of feeding. The authors compared the risks of pneumonitis and other important outcomes associated with small intestinal and gastric feeding in patients with severe TBI. METHODS This systematic review and meta-analysis was conducted according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. Relevant randomized controlled trials (up to December 16, 2013) that compared small bowel to gastric feeding in patients with severe TBI were identified from searches in the PubMed and Embase databases. The primary outcome was risk of pneumonia. Secondary outcomes included ventilator-associated pneumonia, mortality, length of intensive care unit stay, length of hospital stay, duration of mechanical ventilation, total number of complications, aspiration, diarrhea, distention, Glasgow Coma Scale score, Injury Severity Score, and Acute Physiology and Chronic Health Evaluation II score. RESULTS Five randomized controlled trials with 325 participants in total were included in the meta-analysis. Compared with gastric feeding, small bowel feeding was associated with a significant reduction in the incidence of pneumonitis (risk ratio [RR] 0.67; 95% CI 0.52-0.87; p=0.002; I2=0.0%) and ventilator-associated pneumonia (RR 0.52; 95% CI 0.34-0.81; p=0.003; I2=0.0%). Small intestinal feeding was also associated with a decrease in the total number of complications (RR 0.43; 95% CI 0.20-0.93; p=0.03; I2=68%). However, small intestinal feeding did not seem to significantly convert any of the other end points in the meta-analysis. CONCLUSIONS The limited evidence suggests that small bowel feeding in patients with severe TBI is associated with a risk of pneumonia that is lower than that with gastric feeding. From this result, the authors recommend the use of small intestinal feeding to reduce the incidence of pneumonitis in patients with severe TBI.
Collapse
Affiliation(s)
- Dong Wang
- Department of Neurosurgery, The First Affiliated Hospital of Shantou University Medical College
- Department of Histology and Embryology, Shantou University Medical College, Shantou, Guangdong, China; and
| | - Shao-Qin Zheng
- Department of Neurosurgery, The First Affiliated Hospital of Shantou University Medical College
| | - Xian-Cai Chen
- Department of Histology and Embryology, Shantou University Medical College, Shantou, Guangdong, China; and
| | - Shi-Wen Jiang
- Department of Biomedical Science, Mercer University School of Medicine, Savannah, Georgia
| | - Hai-Bin Chen
- Department of Neurosurgery, The First Affiliated Hospital of Shantou University Medical College
- Department of Histology and Embryology, Shantou University Medical College, Shantou, Guangdong, China; and
| |
Collapse
|
119
|
Pierre JF, Busch RA, Kudsk KA. The gastrointestinal immune system: Implications for the surgical patient. Curr Probl Surg 2015; 53:11-47. [PMID: 26699624 DOI: 10.1067/j.cpsurg.2015.10.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 10/13/2015] [Indexed: 12/27/2022]
Affiliation(s)
- Joseph F Pierre
- Department of Medicine, Section of Gastroenterology, Hepatology, and Nutrition, University of Chicago, Chicago, IL
| | - Rebecca A Busch
- Department of Surgery, Division of General Surgery, University of Wisconsin-Madison, Madison, WI
| | - Kenneth A Kudsk
- Department of Surgery, Division of General Surgery, University of Wisconsin-Madison, Madison, WI; Veterans Administration Surgical Services, William S. Middleton Memorial Veterans Hospital, Madison, WI.
| |
Collapse
|
120
|
Dickerson RN, Voss JR, Schroeppel TJ, Maish GO, Magnotti LJ, Minard G, Croce MA. Feasibility of jejunal enteral nutrition for patients with severe duodenal injuries. Nutrition 2015; 32:309-14. [PMID: 26704967 DOI: 10.1016/j.nut.2015.08.026] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Revised: 08/26/2015] [Accepted: 08/26/2015] [Indexed: 02/08/2023]
Abstract
OBJECTIVE The aim of this study was to evaluate the feasibility of enteral nutrition (EN) for critically ill trauma patients with severe traumatic duodenal injuries who received placement of concurrent decompressing and feeding jejunostomies. METHODS Adult patients admitted to the trauma intensive care unit from January 2010 to December 2013, given concurrent afferent decompressing and efferent feeding jejunostomies for severe duodenal injury and provided EN or parenteral nutrition (PN), were retrospectively evaluated. Enteral feeding intolerance was defined as an increase in the decompressing jejunostomy drainage volume output, worsening abdominal distension, or cramping/pain unrelated to surgical incisions. Patients who failed initial EN were transitioned to PN. RESULTS Twenty-six patients were enrolled. Of the 24 patients given EN within the first 2 wk posthospitalization, 18 (75%) failed EN within 2 ± 2 d of initiating EN. EN was discontinued when increases were seen in decompressing jejunostomy drainage volume output (n = 11) and output with abdominal pain and/or distension (n = 6), or abdominal pain/distension was seen without an increase in output (n = 1). Jejunostomy drainage volume output increased from 474 ± 425 mL/d to 1168 ± 725 mL/d (P < 0.001) during EN intolerance. More patients with blunt intestinal injury than those with penetrating injuries (75% versus 15%, respectively; P = 0.035) tolerated EN. Patients initially given PN (n = 13) received more calories (P < 0.005) and protein (P < 0.001) than those given initial EN (n = 13). CONCLUSION The majority of patients with severe duodenal injuries and concurrent decompressing/feeding tube jejunostomies failed initial EN therapy.
Collapse
Affiliation(s)
- Roland N Dickerson
- Department of Clinical Pharmacy, University of Tennessee Health Science Center, Memphis, TN.
| | - Johnathan R Voss
- Department of Clinical Pharmacy, University of Tennessee Health Science Center, Memphis, TN
| | - Thomas J Schroeppel
- Department of Surgery, University of Tennessee Health Science Center, Memphis, TN
| | - George O Maish
- Department of Surgery, University of Tennessee Health Science Center, Memphis, TN
| | - Louis J Magnotti
- Department of Surgery, University of Tennessee Health Science Center, Memphis, TN
| | - Gayle Minard
- Department of Surgery, University of Tennessee Health Science Center, Memphis, TN
| | - Martin A Croce
- Department of Surgery, University of Tennessee Health Science Center, Memphis, TN
| |
Collapse
|
121
|
Gut Lymphocyte Phenotype Changes After Parenteral Nutrition and Neuropeptide Administration. Ann Surg 2015; 262:194-201. [PMID: 25563877 DOI: 10.1097/sla.0000000000000878] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECTIVE To define gut-associated lymphoid tissue (GALT) phenotype changes with parenteral nutrition (PN) and PN with bombesin (BBS). BACKGROUND PN reduces respiratory tract (RT) and GALT Peyer patch and lamina propria lymphocytes, lowers gut and RT immunoglobulin A (IgA) levels, and destroys established RT antiviral and antibacterial immunity. BBS, an enteric nervous system neuropeptide, reverses PN-induced IgA and RT immune defects. METHODS Experiment 1: Intravenously cannulated ICR mice received chow, PN, or PN + BBS injections for 5 days. LSR-II flow cytometer analyzed Peyer patches and lamina propria isolated lymphocytes for homing phenotypes (L-selectin and LPAM-1) and state of activation (CD25, CD44) in T (CD3)-cell subsets (CD4 and CD8) along with homing phenotype (L-selectin and LPAM-1) in naive B (IgD) and antigen-activated (IgD or IgM) B (CD45R/B220) cells. Experiment 2: Following the initial experiment 1 protocol, lamina propria T regulatory cell phenotype was evaluated by Foxp3 expression. RESULTS Experiment 1: PN significantly reduced lamina propria (1) CD4CD25 (activated) and (2) CD4CD25LPAM-1 (activated cells homed to the lamina propria) T cells, whereas PN-BBS assimilated chow levels. PN significantly reduced lamina propria (1) IgD (naive), (2) IgDLPAM (antigen-activated homed to the lamina propria) and CD44 memory B cells, whereas PN-BBS assimilated chow levels. Experiment 2: PN significantly reduced lamina propria CD4CD25Foxp3 T regulatory cells compared with chow-fed mice, whereas PN + BBS assimilated chow levels. CONCLUSIONS PN reduces lamina propria activated and T regulatory cells and also naive and memory B cells. BBS addition to PN maintains these cell phenotypes, demonstrating the intimate involvement of the enteric nervous system in mucosal immunity.
Collapse
|
122
|
Partial Enteral Nutrition Preserves Elements of Gut Barrier Function, Including Innate Immunity, Intestinal Alkaline Phosphatase (IAP) Level, and Intestinal Microbiota in Mice. Nutrients 2015; 7:6294-312. [PMID: 26247961 PMCID: PMC4555127 DOI: 10.3390/nu7085288] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Revised: 05/20/2015] [Accepted: 07/22/2015] [Indexed: 01/28/2023] Open
Abstract
Lack of enteral nutrition (EN) during parenteral nutrition (PN) leads to higher incidence of infection because of gut barrier dysfunction. However, the effects of partial EN on intestina linnate immunity, intestinal alkaline phosphatase (IAP) and microbiota remain unclear. The mice were randomized into six groups to receive either standard chow or isocaloric and isonitrogenous nutritional support with variable partial EN to PN ratios. Five days later, the mice were sacrificed and tissue samples were collected. Bacterial translocation, the levels of lysozyme, mucin 2 (MUC2), and IAP were analyzed. The composition of intestinal microbiota was analyzed by 16S rRNA pyrosequencing. Compared with chow, total parenteral nutrition (TPN) resulted in a dysfunctional mucosal barrier, as evidenced by increased bacterial translocation (p < 0.05), loss of lysozyme, MUC2, and IAP, and changes in the gut microbiota (p < 0.001). Administration of 20% EN supplemented with PN significantly increased the concentrations of lysozyme, MUC2, IAP, and the mRNA levels of lysozyme and MUC2 (p < 0.001). The percentages of Bacteroidetes and Tenericutes were significantly lower in the 20% EN group than in the TPN group (p < 0.001). These changes were accompanied by maintained barrier function in bacterial culture (p < 0.05). Supplementation of PN with 20% EN preserves gut barrier function, by way of maintaining innate immunity, IAP and intestinal microbiota.
Collapse
|
123
|
Busch RA, Heneghan AF, Pierre JF, Neuman JC, Reimer CA, Wang X, Kimple ME, Kudsk KA. Bombesin Preserves Goblet Cell Resistin-Like Molecule β During Parenteral Nutrition but Not Other Goblet Cell Products. JPEN J Parenter Enteral Nutr 2015; 40:1042-9. [PMID: 25934045 DOI: 10.1177/0148607115585353] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 04/07/2015] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Parenteral nutrition (PN) increases the risk of infection in critically ill patients and is associated with defects in gastrointestinal innate immunity. Goblet cells produce mucosal defense compounds, including mucin (principally MUC2), trefoil factor 3 (TFF3), and resistin-like molecule β (RELMβ). Bombesin (BBS), a gastrin-releasing peptide analogue, experimentally reverses PN-induced defects in Paneth cell innate immunity. We hypothesized that PN reduces goblet cell product expression and PN+BBS would reverse these PN-induced defects. METHODS Two days after intravenous cannulation, male Institute of Cancer Research mice were randomized to chow (n = 15), PN (n = 13), or PN+BBS (15 µg tid) (n = 12) diets for 5 days. Defined segments of ileum and luminal fluid were analyzed for MUC2, TFF3, and RELMβ by quantitative reverse transcriptase polymerase chain reaction and Western blot. Th2 cytokines interleukin (IL)-4 and IL-13 were measured by enzyme-linked immunosorbent assay. RESULTS Compared with chow, PN significantly reduced MUC2 in ileum (P < .01) and luminal fluid (P = .01). BBS supplementation did not improve ileal or luminal MUC2 compared with PN (P > .3). Compared with chow, PN significantly reduced TFF3 in ileum (P < .02) and luminal fluid (P < .01). BBS addition did not improve ileal or luminal TFF3 compared with PN (P > .3). Compared with chow, PN significantly reduced ileal RELMβ (P < .01). BBS supplementation significantly increased ileal RELMβ to levels similar to chow (P < .03 vs PN; P > .6 vs chow). Th2 cytokines were decreased with PN and returned to chow levels with BBS. CONCLUSION PN significantly impairs the goblet cell component of innate mucosal immunity. BBS only preserves goblet cell RELMβ during PN but not other goblet cell products measured.
Collapse
Affiliation(s)
- Rebecca A Busch
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Aaron F Heneghan
- Veteran Administration Surgical Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Joseph F Pierre
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA Department of Medicine, Division of Gastroenterology, University of Chicago, Chicago, Illinois, USA
| | - Joshua C Neuman
- Department of Nutritional Sciences, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Claire A Reimer
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Xinying Wang
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA Department of Surgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Michelle E Kimple
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Kenneth A Kudsk
- Veteran Administration Surgical Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| |
Collapse
|
124
|
Gong L, Yan B, Chen Y, Wang M, Zhang Q, Hui C, Wang C. Alternative method for jejunostomy in Ivor-Lewis esophagectomy. Thorac Cancer 2015; 6:296-302. [PMID: 26273375 PMCID: PMC4448396 DOI: 10.1111/1759-7714.12182] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 09/15/2014] [Indexed: 12/18/2022] Open
Abstract
Background To supplement nutrition, jejunostomy has been widely adopted as an adjunct surgical procedure for Ivor-Lewis esophagectomy. Most Chinese surgeons have a preference for parenteral nutrition even though it has some disadvantages compared with jejunostomy. In this report, we describe a new approach that allows the quick insertion of a feeding tube in Ivor-Lewis esophagectomy. We retrospectively analyze cases that have applied this approach and compare the advantages and disadvantages of jejunostomy. Methods Between January 2010 and December 2012, 131 patients underwent Ivor-Lewis esophagectomy in our hospital. These patients were divided into three groups: the total parenteral nutrition (PN) group, the jejunostomy (JT) group and the feeding tube (FT) group. The effect and safety of the procedure were compared. Results It took approximately 20 minutes longer to perform jejunostomy compared to placing a feeding tube (P < 0.05). The nutrition cost of the JT group was higher than the FT group (P < 0.05). There was no significant difference between the FT and JT groups (P > 0.05) in the ratio of body weight loss seven days post-surgery. The anal exsufflation time of the FT group was similar to the JT group (P > 0.05). The incidence of intestinal adhesion and obstruction in the JT group was 26.3%, which is much higher than in the FT and PN groups (P < 0.05). Conclusion Placing the feeding tube after Ivor-Lewis esophagectomy can decrease operative damage and bring sufficient nutrition. We believe it can be an alternative to jejunostomy in Ivor-Lewis esophagectomy.
Collapse
Affiliation(s)
- Liqun Gong
- Department of Lung Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer Tianjin, China ; Key Laboratory of Cancer Prevention and Therapy Tianjin, China
| | - Bo Yan
- Department of Radiotherapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer Tianjin, China
| | - Yulong Chen
- Department of Lung Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer Tianjin, China ; Key Laboratory of Cancer Prevention and Therapy Tianjin, China
| | - Meng Wang
- Department of Lung Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer Tianjin, China ; Key Laboratory of Cancer Prevention and Therapy Tianjin, China
| | - Qiang Zhang
- Department of Lung Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer Tianjin, China ; Key Laboratory of Cancer Prevention and Therapy Tianjin, China
| | - Chen Hui
- Department of Lung Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer Tianjin, China ; Key Laboratory of Cancer Prevention and Therapy Tianjin, China
| | - Changli Wang
- Department of Lung Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer Tianjin, China ; Key Laboratory of Cancer Prevention and Therapy Tianjin, China
| |
Collapse
|
125
|
Freeman JJ, Feng Y, Demehri FR, Dempsey PJ, Teitelbaum DH. TPN-associated intestinal epithelial cell atrophy is modulated by TLR4/EGF signaling pathways. FASEB J 2015; 29:2943-58. [PMID: 25782989 DOI: 10.1096/fj.14-269480] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 03/02/2015] [Indexed: 12/12/2022]
Abstract
Recent studies suggest a close interaction between epidermal growth factor (EGF) and TLR signaling in the modulation of intestinal epithelial cell (IEC) proliferation; however, how these signaling pathways adjust IEC proliferation is poorly understood. We utilized a model of total parenteral nutrition (TPN), or enteral nutrient deprivation, to study this interaction as TPN results in mucosal atrophy due to decreased IEC proliferation and increased apoptosis. We identified the novel finding of decreased mucosal atrophy in TLR4 knockout (TLR4KO) mice receiving TPN. We hypothesized that EGF signaling is preserved in TLR4KO-TPN mice and prevents mucosal atrophy. C57Bl/6 and strain-matched TLR4KO mice were provided either enteral feeding or TPN. IEC proliferation and apoptosis were measured. Cytokine and growth factor abundances were detected in both groups. To examine interdependence of these pathways, ErbB1 pharmacologic blockade was used. The marked decline in IEC proliferation with TPN was nearly prevented in TLR4KO mice, and intestinal length was partially preserved. EGF was significantly increased, and TNF-α decreased in TLR4KO-TPN versus wild-type (WT)-TPN mice. Apoptotic positive crypt cells were 15-fold higher in WT-TPN versus TLR4KO-TPN mice. Bcl-2 was significantly increased in TLR4KO-TPN mice, while Bax decreased 10-fold. ErbB1 blockade prevented this otherwise protective effect in TLR4KO-sTPN mice. TLR4 blockade significantly prevented TPN-associated atrophy by preserving proliferation and preventing apoptosis. This is driven by a reduction in TNF-α abundance and increased EGF. Potential manipulation of this regulatory pathway may have significant clinical potential to prevent TPN-associated atrophy.
Collapse
Affiliation(s)
- Jennifer J Freeman
- *Department of Surgery, Section of Pediatric Surgery, and Center for Organogenesis, University of Michigan, Ann Arbor, Michigan, USA; and School of Medicine, Department of Pediatrics, University of Colorado, Denver, Anschutz Medical Campus, Denver, Colorado, USA
| | - Yongjia Feng
- *Department of Surgery, Section of Pediatric Surgery, and Center for Organogenesis, University of Michigan, Ann Arbor, Michigan, USA; and School of Medicine, Department of Pediatrics, University of Colorado, Denver, Anschutz Medical Campus, Denver, Colorado, USA
| | - Farokh R Demehri
- *Department of Surgery, Section of Pediatric Surgery, and Center for Organogenesis, University of Michigan, Ann Arbor, Michigan, USA; and School of Medicine, Department of Pediatrics, University of Colorado, Denver, Anschutz Medical Campus, Denver, Colorado, USA
| | - Peter J Dempsey
- *Department of Surgery, Section of Pediatric Surgery, and Center for Organogenesis, University of Michigan, Ann Arbor, Michigan, USA; and School of Medicine, Department of Pediatrics, University of Colorado, Denver, Anschutz Medical Campus, Denver, Colorado, USA
| | - Daniel H Teitelbaum
- *Department of Surgery, Section of Pediatric Surgery, and Center for Organogenesis, University of Michigan, Ann Arbor, Michigan, USA; and School of Medicine, Department of Pediatrics, University of Colorado, Denver, Anschutz Medical Campus, Denver, Colorado, USA
| |
Collapse
|
126
|
Ralls MW, Demehri FR, Feng Y, Woods Ignatoski KM, Teitelbaum DH. Enteral nutrient deprivation in patients leads to a loss of intestinal epithelial barrier function. Surgery 2015; 157:732-42. [PMID: 25704423 DOI: 10.1016/j.surg.2014.12.004] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Revised: 11/17/2014] [Accepted: 12/03/2014] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To investigate the effect of nutrient withdrawal on human intestinal epithelial barrier function (EBF). We hypothesized that unfed mucosa results in decreased EBF. This was tested in a series of surgical small intestinal resection specimens. DESIGN Small bowel specifically excluding inflamed tissue, was obtained from pediatric patients (aged 2 days to 19 years) undergoing intestinal resection. EBF was assessed in Ussing chambers for transepithelial resistance (TER) and passage of fluorescein isothiocyanate (FITC)-dextran (4 kD). Tight junction and adherence junction proteins were imaged with immunofluorescence staining. Expression of Toll-like receptors (TLR) and inflammatory cytokines were measured in loop ileostomy takedowns in a second group of patients. RESULTS Because TER increased with patient age (P < .01), results were stratified into infant versus teenage groups. Fed bowel had significantly greater TER versus unfed bowel (P < .05) in both age populations. Loss of EBF was also observed by an increase in FITC-dextran permeation in enteral nutrient-deprived segments (P < .05). Immunofluorescence staining showed marked declines in intensity of ZO-1, occludin, E-cadherin, and claudin-4 in unfed intestinal segments, as well as a loss of structural formation of tight junctions. Analysis of cytokine and TLR expression showed significant increases in tumor necrosis factor (TNF)-α and TLR4 in unfed segments of bowel compared with fed segments from the same individual. CONCLUSION EBF declined in unfed segments of human small bowel. This work represents the first direct examination of EBF from small bowel derived from nutrient-deprived humans and may explain the increased incidence of infectious complications seen in patients not receiving enteral feeds.
Collapse
Affiliation(s)
- Matthew W Ralls
- Section of Pediatric Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI
| | - Farokh R Demehri
- Section of Pediatric Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI
| | - Yongjia Feng
- Section of Pediatric Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI
| | | | - Daniel H Teitelbaum
- Section of Pediatric Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI.
| |
Collapse
|
127
|
Rosenthal MD, Vanzant EL, Martindale RG, Moore FA. Evolving paradigms in the nutritional support of critically ill surgical patients. Curr Probl Surg 2015; 52:147-82. [PMID: 25946621 DOI: 10.1067/j.cpsurg.2015.02.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 01/29/2015] [Accepted: 02/11/2015] [Indexed: 12/12/2022]
|
128
|
The enteric nervous system neuropeptide, bombesin, reverses innate immune impairments during parenteral nutrition. Ann Surg 2015; 260:432-43; discussion 443-4. [PMID: 25115419 DOI: 10.1097/sla.0000000000000871] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Lack of enteral stimulation during parenteral nutrition (PN) impairs mucosal immunity. Bombesin (BBS), a gastrin-releasing peptide analogue, reverses PN-induced defects in acquired immunity. Paneth cells produce antimicrobial peptides (AMPs) of innate immunity for release after cholinergic stimulation. OBJECTIVE Determine if BBS restores AMPs and bactericidal function during PN. METHODS Intravenously cannulated male ICR mice were randomized to Chow, PN, or PN+BBS (15 μg 3 times daily, n = 7 per group) for 5 days. Ileum was analyzed for AMPs (Protein: sPLA2 by fluorescence, lysozyme and RegIII-γ by western andcryptdin-4 by ELISA; mRNA: all by RT-PCR). Cholinergic stimulated (100 μM bethanechol) ileal specimens assessed Pseudomonas bactericidal activity. Ileum (Chow: n = 7; PN: n = 9; PN+BBS: n = 8) was assessed for Escherichia coli invasion in ex-vivo culture. RESULTS PN significantly decreased most AMPs versus Chow while BBS maintained Chow levels (sPLA2: Chow: 107 + 14*, PN: 44.6 + 7.2, PN+BBS: 78.7 + 13.4* Fl/min/μL/total protein; Lysozyme: Chow: 63.9 + 11.9*, PN: 26.8 + 6.2; PN+BBS: 64.9 + 13.8* lysozyme/total protein; RegIII-γ: Chow: 51.5 + 10.0*, PN: 20.4 + 4.3, PN+BBS: 31.0 + 8.4 RegIII-γ/total protein; Cryptdin-4: Chow: 18.4 + 1.5*, PN: 12.7 + 1.6, PN+BBS: 26.1 + 2.4*† pg/mg [all *P < 0.05 vs PN and †P < 0.05 vs Chow]). Functionally, BBS prevented PN loss of bactericidal activity after cholinergic stimulation (Chow: 25.3 + 3.6*, PN: 13.0 + 3.2; PN+BBS: 27.0 + 4.7* percent bacterial killing, *P < 0.05 vs PN). BBS reduced bacterial invasion in unstimulated tissue barely missing significance (P = 0.06). CONCLUSIONS The enteric nervous system (ENS) controls AMP levels in Paneth cells during PN but mucosal protection by innate immunity requires both ENS and parasympathetic stimulation.
Collapse
|
129
|
Linn DD, Beckett RD, Foellinger K. Administration of enteral nutrition to adult patients in the prone position. Intensive Crit Care Nurs 2015; 31:38-43. [DOI: 10.1016/j.iccn.2014.07.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Revised: 07/09/2014] [Accepted: 07/12/2014] [Indexed: 01/20/2023]
|
130
|
Yin J, Wang J, Zhang S, Yao D, Mao Q, Kong W, Ren L, Li Y, Li J. Early versus delayed enteral feeding in patients with abdominal trauma: a retrospective cohort study. Eur J Trauma Emerg Surg 2015; 41:99-105. [PMID: 26038172 DOI: 10.1007/s00068-014-0425-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Accepted: 06/02/2014] [Indexed: 10/25/2022]
Abstract
PURPOSE Early enteral feeding within 24-48 h of intensive care unit admission is recommended for critically ill patients. This study aimed to determine if early enteral feeding could be safely implemented with purported benefits in patients with abdominal trauma. METHODS A retrospective cohort study was performed that included 88 adult patients with abdominal trauma. Patients receiving enteral feeding within 72 h of surgical intensive care unit (SICU) admission (early-initiation group, n = 28) were compared to those receiving enteral feeding later (delayed-initiation group, n = 60). RESULTS The two groups were comparable in demographic characteristics and injury severity. There were no differences in feeding intolerance (53.6 vs. 43.3%, p = 0.37) and mortality at 28 days (0 vs. 5%, p = 0.55) between the early-initiation group and the delayed-initiation group. However, patients in the early-initiation group had fewer infectious complications (17.9 vs. 40 %, p = 0.04) and shorter length of stay in SICU and hospital (p < 0.01) than patients in the delayed-initiation group. CONCLUSIONS Early enteral feeding administered within 72 h of SICU admission was associated with improved clinical outcomes without risk of increasing feeding intolerance in patients with abdominal trauma. Our results support the implementation of early enteral feeding in abdominal trauma management.
Collapse
Affiliation(s)
- Jianyi Yin
- Department of Surgery, Jinling Hospital, Nanjing University School of Medicine, 305 East Zhongshan Road, Nanjing, 210002, China
| | | | | | | | | | | | | | | | | |
Collapse
|
131
|
Lee SH, Jang JY, Kim HW, Jung MJ, Lee JG. Effects of early enteral nutrition on patients after emergency gastrointestinal surgery: a propensity score matching analysis. Medicine (Baltimore) 2014; 93:e323. [PMID: 25526487 PMCID: PMC4603119 DOI: 10.1097/md.0000000000000323] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Revised: 11/07/2014] [Accepted: 11/10/2014] [Indexed: 01/24/2023] Open
Abstract
Early postoperative enteral feeding has been demonstrated to improve the outcome of patients who underwent surgery for gastrointestinal (GI) malignancies, trauma, perforation, and/or obstruction. Thus, this study was conducted to assess the efficacy of early postoperative enteral nutrition (EN) after emergency surgery in patients with GI perforation or strangulation. The medical records of 484 patients, admitted between January 2007 and December 2012, were reviewed retrospectively. Patients were divided into 2 groups: the early EN (EEN, N=77) group and the late EN (LEN, N=407) group. The morbidity, mortality, length of hospital, and intensive care unit (ICU) stays were compared between the 2 groups. Propensity score matching was performed in order to adjust for any baseline differences. Patients receiving EEN had reduced in-hospital mortality rates (EEN 4.5% vs LEN 19.4%; P=0.008), pulmonary complications (EEN 4.5% vs LEN 19.4%; P=0.008), lengths of hospital stay (median: 14.0, interquartile range: 8.0-24.0 vs median: 17.0, interquartile range: 11.0-26.0, P=0.048), and more 28-day ICU-free days (median: 27.0, interquartile range: 25.0-27.0 vs median: 25.0, interquartile range: 22.0-27.0, P=0.042) than those receiving LEN in an analysis using propensity score matching. The significant difference in survival between the 2 groups was also shown in the Kaplan-Meier survival curve (P=0.042). In a further analysis using the Cox proportional hazard ratio after matching on the propensity score, EEN was associated with reduced in-hospital mortality (hazard ratio, 0.03; 95% confidence interval, 0.01-0.49; P=0.015). EEN is associated with beneficial effects, such as reduced in-hospital mortality rates, pulmonary complications, lengths of hospital stay, and more 28-day ICU-free days, after emergency GI surgery.
Collapse
Affiliation(s)
- Seung Hwan Lee
- From the Department of Surgery (SHL, HWK, MJJ, JGL), Yonsei University College of Medicine, Seoul; and Department of Surgery (JYJ), Yonsei University Wonju College of Medicine, Wonju, Korea
| | | | | | | | | |
Collapse
|
132
|
Busch RA, Jonker MA, Pierre JF, Heneghan AF, Kudsk KA. Innate Mucosal Immune System Response of BALB/c vs C57BL/6 Mice to Injury in the Setting of Enteral and Parenteral Feeding. JPEN J Parenter Enteral Nutr 2014; 40:256-63. [PMID: 25403938 DOI: 10.1177/0148607114558489] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 10/13/2014] [Indexed: 11/15/2022]
Abstract
BACKGROUND Outbred mice exhibit increased airway and intestinal immunoglobulin A (IgA) following injury when fed normal chow, consistent with humans. Parenteral nutrition (PN) eliminates IgA increases at both sites. Inbred mice are needed for detailed immunological studies; however, specific strains have not been evaluated for this purpose. BALB/c and C57BL/6 are common inbred mouse strains but demonstrate divergent immune responses to analogous stress. This study addressed which inbred mouse strain best replicates the outbred mouse and human immune response to injury. METHODS Intravenously cannulated mice received chow or PN for 5 days and then underwent sacrifice at 0 or 8 hours following controlled surgical injury (BALB/c: n = 16-21/group; C57BL/6: n = 12-15/group). Bronchoalveolar lavage (BAL) was analyzed by enzyme-linked immunosorbent assay for IgA, tumor necrosis factor-α (TNF-α), interleukin (IL)-1β, and IL-6, while small intestinal wash fluid (SIWF) was analyzed for IgA. RESULTS No significant increase in BAL IgA occurred following injury in chow- or PN-fed BALB/c mice (chow: P = .1; PN: P = .7) despite significant increases in BAL TNF-α and SIWF IgA (chow: 264 ± 28 vs 548 ± 37, P < .0001; PN: 150 ± 12 vs 301 ± 17, P < .0001). Injury significantly increased mucosal IgA in chow-fed C57BL/6 mice (BAL: 149 ± 33 vs 342 ± 87, P = .01; SIWF: 236 ± 28 vs 335 ± 32, P = .006) and BAL cytokines. After injury, PN-fed C57BL/6 mice exhibited no difference in BAL IgA (P = .9), BAL cytokines, or SIWF IgA (P = .1). CONCLUSIONS C57BL/6 mice exhibit similar airway responses to injury as outbred mice and humans, providing an appropriate model for studying mucosal responses to injury. The BALB/c mucosal immune system responds differently to injury and does not replicate the human injury response.
Collapse
Affiliation(s)
- Rebecca A Busch
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Mark A Jonker
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Joseph F Pierre
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin Department of Medicine-Gastroenterology, University of Chicago, Chicago, Illinois
| | - Aaron F Heneghan
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Kenneth A Kudsk
- Veterans Administration Surgical Services, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| |
Collapse
|
133
|
Charles EJ, Petroze RT, Metzger R, Hranjec T, Rosenberger LH, Riccio LM, McLeod MD, Guidry CA, Stukenborg GJ, Swenson BR, Willcutts KF, O'Donnell KB, Sawyer RG. Hypocaloric compared with eucaloric nutritional support and its effect on infection rates in a surgical intensive care unit: a randomized controlled trial. Am J Clin Nutr 2014; 100:1337-43. [PMID: 25332331 PMCID: PMC4196484 DOI: 10.3945/ajcn.114.088609] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Proper caloric intake goals in critically ill surgical patients are unclear. It is possible that overnutrition can lead to hyperglycemia and an increased risk of infection. OBJECTIVE This study was conducted to determine whether surgical infection outcomes in the intensive care unit (ICU) could be improved with the use of hypocaloric nutritional support. DESIGN Eighty-three critically ill patients were randomly allocated to receive either the standard calculated daily caloric requirement of 25-30 kcal · kg(-1) · d(-1) (eucaloric) or 50% of that value (hypocaloric) via enteral tube feeds or parenteral nutrition, with an equal protein allocation in each group (1.5 g · kg(-1) · d(-1)). RESULTS There were 82 infections in the hypocaloric group and 66 in the eucaloric group, with no significant difference in the mean (± SE) number of infections per patient (2.0 ± 0.6 and 1.6 ± 0.2, respectively; P = 0.50), percentage of patients acquiring infection [70.7% (29 of 41) and 76.2% (32 of 42), respectively; P = 0.57], mean ICU length of stay (16.7 ± 2.7 and 13.5 ± 1.1 d, respectively; P = 0.28), mean hospital length of stay (35.2 ± 4.9 and 31.0 ± 2.5 d, respectively; P = 0.45), mean 0600 glucose concentration (132 ± 2.9 and 135 ± 3.1 mg/dL, respectively; P = 0.63), or number of mortalities [3 (7.3%) and 4 (9.5%), respectively; P = 0.72]. Further analyses revealed no differences when analyzed by sex, admission diagnosis, site of infection, or causative organism. CONCLUSIONS Among critically ill surgical patients, caloric provision across a wide acceptable range does not appear to be associated with major outcomes, including infectious complications. The optimum target for caloric provision remains elusive.
Collapse
Affiliation(s)
- Eric J Charles
- From the Department of Surgery, University of Virginia Health System, Charlottesville, VA (EJC, RTP, RM, TH, LHR, LMR, MDM, CAG, BRS, KFW, KBO, and RGS), and the Department of Public Health Sciences, University of Virginia School of Medicine, Charlottesville, VA (GJS)
| | - Robin T Petroze
- From the Department of Surgery, University of Virginia Health System, Charlottesville, VA (EJC, RTP, RM, TH, LHR, LMR, MDM, CAG, BRS, KFW, KBO, and RGS), and the Department of Public Health Sciences, University of Virginia School of Medicine, Charlottesville, VA (GJS)
| | - Rosemarie Metzger
- From the Department of Surgery, University of Virginia Health System, Charlottesville, VA (EJC, RTP, RM, TH, LHR, LMR, MDM, CAG, BRS, KFW, KBO, and RGS), and the Department of Public Health Sciences, University of Virginia School of Medicine, Charlottesville, VA (GJS)
| | - Tjasa Hranjec
- From the Department of Surgery, University of Virginia Health System, Charlottesville, VA (EJC, RTP, RM, TH, LHR, LMR, MDM, CAG, BRS, KFW, KBO, and RGS), and the Department of Public Health Sciences, University of Virginia School of Medicine, Charlottesville, VA (GJS)
| | - Laura H Rosenberger
- From the Department of Surgery, University of Virginia Health System, Charlottesville, VA (EJC, RTP, RM, TH, LHR, LMR, MDM, CAG, BRS, KFW, KBO, and RGS), and the Department of Public Health Sciences, University of Virginia School of Medicine, Charlottesville, VA (GJS)
| | - Lin M Riccio
- From the Department of Surgery, University of Virginia Health System, Charlottesville, VA (EJC, RTP, RM, TH, LHR, LMR, MDM, CAG, BRS, KFW, KBO, and RGS), and the Department of Public Health Sciences, University of Virginia School of Medicine, Charlottesville, VA (GJS)
| | - Matthew D McLeod
- From the Department of Surgery, University of Virginia Health System, Charlottesville, VA (EJC, RTP, RM, TH, LHR, LMR, MDM, CAG, BRS, KFW, KBO, and RGS), and the Department of Public Health Sciences, University of Virginia School of Medicine, Charlottesville, VA (GJS)
| | - Christopher A Guidry
- From the Department of Surgery, University of Virginia Health System, Charlottesville, VA (EJC, RTP, RM, TH, LHR, LMR, MDM, CAG, BRS, KFW, KBO, and RGS), and the Department of Public Health Sciences, University of Virginia School of Medicine, Charlottesville, VA (GJS)
| | - George J Stukenborg
- From the Department of Surgery, University of Virginia Health System, Charlottesville, VA (EJC, RTP, RM, TH, LHR, LMR, MDM, CAG, BRS, KFW, KBO, and RGS), and the Department of Public Health Sciences, University of Virginia School of Medicine, Charlottesville, VA (GJS)
| | - Brian R Swenson
- From the Department of Surgery, University of Virginia Health System, Charlottesville, VA (EJC, RTP, RM, TH, LHR, LMR, MDM, CAG, BRS, KFW, KBO, and RGS), and the Department of Public Health Sciences, University of Virginia School of Medicine, Charlottesville, VA (GJS)
| | - Kate F Willcutts
- From the Department of Surgery, University of Virginia Health System, Charlottesville, VA (EJC, RTP, RM, TH, LHR, LMR, MDM, CAG, BRS, KFW, KBO, and RGS), and the Department of Public Health Sciences, University of Virginia School of Medicine, Charlottesville, VA (GJS)
| | - Kelly B O'Donnell
- From the Department of Surgery, University of Virginia Health System, Charlottesville, VA (EJC, RTP, RM, TH, LHR, LMR, MDM, CAG, BRS, KFW, KBO, and RGS), and the Department of Public Health Sciences, University of Virginia School of Medicine, Charlottesville, VA (GJS)
| | - Robert G Sawyer
- From the Department of Surgery, University of Virginia Health System, Charlottesville, VA (EJC, RTP, RM, TH, LHR, LMR, MDM, CAG, BRS, KFW, KBO, and RGS), and the Department of Public Health Sciences, University of Virginia School of Medicine, Charlottesville, VA (GJS)
| |
Collapse
|
134
|
Oda S, Aibiki M, Ikeda T, Imaizumi H, Endo S, Ochiai R, Kotani J, Shime N, Nishida O, Noguchi T, Matsuda N, Hirasawa H. The Japanese guidelines for the management of sepsis. J Intensive Care 2014; 2:55. [PMID: 25705413 PMCID: PMC4336273 DOI: 10.1186/s40560-014-0055-2] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 09/16/2014] [Indexed: 02/08/2023] Open
Abstract
This is a guideline for the management of sepsis, developed by the Sepsis Registry Committee of The Japanese Society of Intensive Care Medicine (JSICM) launched in March 2007. This guideline was developed on the basis of evidence-based medicine and focuses on unique treatments in Japan that have not been included in the Surviving Sepsis Campaign guidelines (SSCG), as well as treatments that are viewed differently in Japan and in Western countries. Although the methods in this guideline conform to the 2008 SSCG, the Japanese literature and the results of the Sepsis Registry Survey, which was performed twice by the Sepsis Registry Committee in intensive care units (ICUs) registered with JSICM, are also referred. This is the first and original guideline for sepsis in Japan and is expected to be properly used in daily clinical practice. This article is translated from Japanese, originally published as “The Japanese Guidelines for the Management of Sepsis” in the Journal of the Japanese Society of Intensive Care Medicine (J Jpn Soc Intensive Care Med), 2013; 20:124–73. The original work is at http://dx.doi.org/10.3918/jsicm.20.124.
Collapse
Affiliation(s)
- Shigeto Oda
- Department of Emergency and Critical Care Medicine, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-Ku, Chiba 260-8677 Japan
| | - Mayuki Aibiki
- Department of Emergency Medicine, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime 791-0295 Japan
| | - Toshiaki Ikeda
- Division of Critical Care and Emergency Medicine, Tokyo Medical University Hachioji Medical Center, 1163 Tatemachi, Hachioji, Tokyo 193-0998 Japan
| | - Hitoshi Imaizumi
- Department of Intensive Care Medicine, Sapporo Medical University School of Medicine, S1 W17, Chuo-ku, Sapporo, 060-8556 Japan
| | - Shigeatsu Endo
- Department of Emergency Medicine, Iwate Medical University, 19-1 Uchimaru, Morioka, Iwate 020-0023 Japan
| | - Ryoichi Ochiai
- First Department of Anesthesia, Toho University School of Medicine, 6-11-1 Omori-nishi, Ota-ku, Tokyo 143-8541 Japan
| | - Joji Kotani
- Department of Emergency, Disaster and Critical Care Medicine, Hyogo College of Medicine, 1-1 Mukogawacho, Nishinomiya, Hyogo 663-8131 Japan
| | - Nobuaki Shime
- Division of Intensive Care Unit, University Hospital, Kyoto Prefectural University of Medicine, Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566 Japan
| | - Osamu Nishida
- Department of Anesthesiology and Critical Care Medicine, Fujita Health University School of Medicine, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi 470-1192 Japan
| | - Takayuki Noguchi
- Department of Anesthesiology and Intensive Care Medicine, Oita University School of Medicine, 1-1 Idaigaoka, Hazamacho, Yufu, Oita 879-5593 Japan
| | - Naoyuki Matsuda
- Emergency and Critical Care Medicine, Graduate School of Medicine Nagoya University, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550 Japan
| | | | | |
Collapse
|
135
|
Bicudo-Salomão A, de Moura RR, de Aguilar-Nascimento JE. Early nutritional therapy in trauma: after A, B, C, D, E, the importance of the F (FEED). Rev Col Bras Cir 2014; 40:342-6. [PMID: 24173487 DOI: 10.1590/s0100-69912013000400015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2012] [Accepted: 08/25/2012] [Indexed: 11/21/2022] Open
Abstract
A significant number of deaths in trauma occurs days to weeks after the initial injury, being caused by infection and organ failure related to hypercatabolism and consequent acute protein malnutrition. Nutritional therapy should be planned and included with other routines of resuscitation for patients with multiple trauma and severe burns. The rapid acquisition of a route for nutritional support is important to start early nutritional therapy within 48 hours of care. The enteral route is the preferred option in traumatized postoperative patients but the parenteral route should be prescribed when enteral feeding is contraindicated or inadequate. After the initial measures dictated by ATLS, synthesized in the A (airway), B (breathing), C (circulation), D (disability) and E (exposure), we include the letter F (feed) to emphasize the importance of early nutritional care in trauma.
Collapse
|
136
|
Panchal AK, Manzi J, Connolly S, Christensen M, Wakeham M, Goday PS, Mikhailov TA. Safety of Enteral Feedings in Critically Ill Children Receiving Vasoactive Agents. JPEN J Parenter Enteral Nutr 2014; 40:236-41. [PMID: 25168592 DOI: 10.1177/0148607114546533] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 07/08/2014] [Indexed: 11/15/2022]
Abstract
BACKGROUND The objective of this retrospective study was to evaluate the safety of enteral feeding in children receiving vasoactive agents (VAs). METHODS Patients aged 1 month to 18 years with a pediatric intensive care unit stay for ≥96 hours during 2007 and 2008 who received any VA (epinephrine, norepinephrine, vasopressin, milrinone, dopamine, and dobutamine) were included and categorized into fed and nonfed groups. Their demographics, clinical characteristics, type and dose of VA, and presence of gastrointestinal (GI) outcomes were obtained. GI outcomes were compared between the groups by the χ(2) test, Mann-Whitney test, and logistic regression. RESULTS In total, 339 patients were included. Of these, 55% were in the fed group and 45% in the nonfed group. Patients in the fed group were younger (median age, 1.05 vs 2.75 years, respectively; P < .001) and tended to have a lower Pediatric Index of Mortality 2 (PIM2) risk of mortality (ROM) than those in the nonfed group (median, 3.33% vs 3.52%, respectively; P = .106). Mortality was lower in the fed group than the nonfed group (6.9% vs 15.9%, respectively; odds ratio [OR], 0.39; 0.18-0.84; P < .01, 95% CI), while GI outcomes did not differ between the groups. The vasoactive-inotropic score (VIS) did not differ between the groups except on day 1 (P = .017). The ROM did not differ between the groups after adjusting for age, PIM2 ROM, and VIS on day 1 (OR, 0.58; 0.26-1.28; P = .18, 95% CI). CONCLUSIONS Enteral feeding in patients receiving VAs is associated with no difference in GI outcomes and a tendency towards lower mortality. Prospective studies are required to confirm the safety of enteral feedings in patients receiving VAs.
Collapse
Affiliation(s)
| | - Jennifer Manzi
- Pediatric Critical Care, Medical College of Wisconsin, Children's Hospital of Wisconsin, Milwaukee, Wisconsin
| | - Susan Connolly
- Pediatric Critical Care, Medical College of Wisconsin, Children's Hospital of Wisconsin, Milwaukee, Wisconsin
| | | | - Martin Wakeham
- Pediatric Critical Care, Medical College of Wisconsin, Children's Hospital of Wisconsin, Milwaukee, Wisconsin
| | - Praveen S Goday
- Pediatric Gastroenterology and Nutrition, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Theresa A Mikhailov
- Pediatric Critical Care, Medical College of Wisconsin, Children's Hospital of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
137
|
Russolillo N, Ferrero A, Vigano' L, Langella S, Briozzo A, Ferlini M, Migliardi M, Capussotti L. Impact of perioperative symbiotic therapy on infectious morbidity after Hpb Surgery in jaundiced patients: a randomized controlled trial. Updates Surg 2014; 66:203-10. [PMID: 25099747 DOI: 10.1007/s13304-014-0259-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Accepted: 07/15/2014] [Indexed: 01/18/2023]
Abstract
This study aimed at evaluating whether the administration of symbiotic therapy in jaundiced patients could reduce their postoperative infectious complications. The study was conducted between November 2008 and February 2011. Jaundiced patients scheduled for elective extrahepatic bile duct resection without liver cirrhosis, intestinal malabsorption or intolerance to symbiotic therapy were randomly assigned to receive [Group A] or not [Group B] symbiotics perioperatively. The primary endpoint was the infectious morbidity rate. Forty patients were included in the analysis (20 in each group). The patients in Group B presented a higher overall morbidity (70 vs 50%) and infectious morbidity rate (50 vs 25%), but the differences were not significant. Eleven patients in Group A (Group ndA) and 13 in Group B (Group ndB) did not receive preoperative biliary drainage. The results of the two groups were comparable. Infectious complications were higher in Group B [5 (34%) vs 0, p = 0.030], while the prevalence of natural killer (NK) cells was higher in Group ndA the day before surgery (17% ± 5.1 vs 10% ± 5.3, p < 0.01) and on post-operative day (POD) 7 (13.1% ± 4.1 vs 7.7% ± 3.4, p < 0.01). The rates of lymph node colonization were similar. The symbiotic therapy failed to reduce the rate of infectious morbidity in jaundiced patients. Further studies investigating the place of symbiotic in no-drainage patients are required.
Collapse
Affiliation(s)
- N Russolillo
- Department of General and Oncological Surgery, 'Umberto I' Mauriziano Hospital, Largo Turati, 62-10128, Turin, Italy,
| | | | | | | | | | | | | | | |
Collapse
|
138
|
Abstract
Providing artificial nutrition is an important part of caring for critically ill patients. However, because of a paucity of robust data, the practice has been highly variable and often based more on dogma than evidence. A number of studies have been published investigating many different aspects of critical care nutrition. Although the influx of data has better informed the practice, the results have often been conflicting or counter to prevailing thought, resulting in discordant opinions and different interpretations by experts in the field. In this article, we review and summarize the data from a number of the published studies, including studies investigating enteral vs parenteral nutrition, supplementing enteral with parenteral nutrition, and use of immunonutrition. In addition, published studies informing the practice of how best to provide enteral nutrition will be reviewed, including the use of trophic feedings, gastric residual volumes, and gastric vs postpyloric tube placement.
Collapse
Affiliation(s)
- Svetang V Desai
- Division of Gastroenterology, Duke University Medical Center, Durham, NC
| | - Stephen A McClave
- Division of Gastroenterology, Hepatology, and Nutrition, University of Louisville School of Medicine, Louisville, KY
| | - Todd W Rice
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, TN.
| |
Collapse
|
139
|
Wang X, Pierre JF, Heneghan AF, Busch RA, Kudsk KA. Glutamine Improves Innate Immunity and Prevents Bacterial Enteroinvasion During Parenteral Nutrition. JPEN J Parenter Enteral Nutr 2014; 39:688-97. [PMID: 24836948 DOI: 10.1177/0148607114535265] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 04/20/2014] [Indexed: 01/08/2023]
Abstract
BACKGROUND Patients receiving parenteral nutrition (PN) are at increased risk of infectious complications compared with enteral feeding, which is in part explained by impaired mucosal immune function during PN. Adding glutamine (GLN) to PN has improved outcome in some clinical patient groups. Although GLN improves acquired mucosal immunity, its effect on innate mucosal immunity (defensins, mucus, lysozymes) has not been investigated. METHODS Forty-eight hours following venous cannulation, male Institute of Cancer Research mice were randomized to chow (n = 10), PN (n = 12), or PN + GLN (n = 13) for 5 days. Small intestine tissue and luminal fluid were collected for mucin 2 (MUC2), lysozyme, cryptdin 4 analysis, and luminal interleukin (IL)-4, IL-10, and IL-13 level measurement. Tissue was also harvested for ex vivo intestinal segment culture to assess tissue susceptibility to enteroinvasive Escherichia coli. RESULTS In both luminal and tissue samples, PN reduced MUC2 and lysozyme (P < .0001, respectively) compared with chow, whereas GLN addition increased MUC2 and lysozyme (luminal, P < .05; tissue, P < .0001, respectively) compared with PN alone. PN significantly suppressed cryptdin 4 expression, while GLN supplementation significantly enhanced expression. IL-4, IL-10, and IL-13 decreased significantly with PN compared with chow, whereas GLN significantly increased these cytokines compared with PN. Functionally, bacterial invasion increased with PN compared with chow (P < .05), while GLN significantly decreased enteroinvasion to chow levels (P < .05). CONCLUSIONS GLN-supplemented PN improves innate immunity and resistance to bacterial mucosal invasion lost with PN alone. This work confirms a clinical rationale for providing glutamine for the protection of the intestinal mucosa.
Collapse
Affiliation(s)
- Xinying Wang
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin Department of Surgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Joseph F Pierre
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin Veterans Administration Surgical Services, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin
| | - Aaron F Heneghan
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin Veterans Administration Surgical Services, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin
| | - Rebecca A Busch
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Kenneth A Kudsk
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin Veterans Administration Surgical Services, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin
| |
Collapse
|
140
|
Lefrant JY, Hurel D, Cano N, Ichai C, Preiser JC, Tamion F. Nutrition artificielle en réanimation. NUTR CLIN METAB 2014. [DOI: 10.1016/j.nupar.2014.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
141
|
Braga M, Wischmeyer PE, Drover J, Heyland DK. Clinical evidence for pharmaconutrition in major elective surgery. JPEN J Parenter Enteral Nutr 2014; 37:66S-72S. [PMID: 24009251 DOI: 10.1177/0148607113494406] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
In recent years, standard nutrition preparations have been modified by adding specific nutrients, such as arginine, ω-3 fatty acids, glutamine, and others, which have been shown to upregulate host immune response, modulate inflammatory response, and improve protein synthesis after surgery. Most randomized trials and several meta-analyses have shown that perioperative administration of enteral arginine, ω-3 fatty acids, and nucleotides (immunonutrition) reduced infection rate and length of hospital stay in patients with upper and lower gastrointestinal (GI) cancer. The most pronounced benefits of immunonutrition were found in subgroups of high-risk and malnourished patients. Promising but not conclusive results have been found in non-GI surgery, especially in head and neck surgery and in cardiac surgery, but larger trials are required before recommending immunonutrition as a routine practice. Conflicting results on the real benefit of parenteral glutamine supplementation in patients undergoing elective major surgery have been published. In conclusion, enteral diets supplemented with specific nutrients significantly improved short-term outcome in patients with cancer undergoing elective GI surgery. Future research should investigate a molecular signaling pathway and identify specific mechanisms of action of immune-enhancing substrates.
Collapse
|
142
|
Jeong O, Ryu SY, Jung MR, Choi WW, Park YK. The safety and feasibility of early postoperative oral nutrition on the first postoperative day after gastrectomy for gastric carcinoma. Gastric Cancer 2014; 17:324-31. [PMID: 23771588 DOI: 10.1007/s10120-013-0275-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2013] [Accepted: 05/23/2013] [Indexed: 02/07/2023]
Abstract
BACKGROUND Unlike the wide acceptance of early enteral nutrition after colorectal surgery, little information is available regarding the feasibility of immediate oral nutrition after gastric cancer surgery. This study evaluated the feasibility and safety of oral nutrition on the first postoperative day after gastrectomy. METHODS From September 2010 to March 2011, 74 consecutive gastric cancer patients received an oral diet on the first postoperative day after gastrectomy. Surgical outcomes, including hospital stay, morbidity, and mortality, were compared with a conventional diet group (n = 96, before September 2010), in which an oral diet was started on the third or fourth postoperative day. RESULTS No significant differences were found in the clinicopathological characteristics or operation types between the two groups. Average diet start times in the early diet (ED) and conventional diet (CD) groups were 1.8 and. 3.2, respectively (p < 0.001). The mean hospital stay was significantly shorter in the ED group (7.4 vs. 8.9 days, p = 0.004). There was no significant difference in postoperative morbidity (p = 0.947) between the two groups. Gastrointestinal-related complications, such as anastomosis leakage or postoperative ileus, were also similar in the two groups. Overall compliance to early oral nutrition in the ED group was 78.5 %, and an old age (≥70 years) was found to affect the compliance to early postoperative oral nutrition. CONCLUSIONS Postoperative oral nutrition is safe and feasible on the first postoperative day after gastrectomy. However, elderly patients require careful monitoring when applying early oral nutrition after gastrectomy.
Collapse
Affiliation(s)
- Oh Jeong
- Division of Gastrointestinal Surgery, Department of Surgery, Chonnam National University Hwasun Hospital, 160 Ilsim-ri, Hwasun-eup, Hwasun-gun, Jeollanam-do, 519-809, South Korea,
| | | | | | | | | |
Collapse
|
143
|
Singer P, Hiesmayr M, Biolo G, Felbinger TW, Berger MM, Goeters C, Kondrup J, Wunder C, Pichard C. Pragmatic approach to nutrition in the ICU: Expert opinion regarding which calorie protein target. Clin Nutr 2014; 33:246-51. [DOI: 10.1016/j.clnu.2013.12.004] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Revised: 12/13/2013] [Accepted: 12/16/2013] [Indexed: 02/06/2023]
|
144
|
Kobayashi L, Coimbra R. Planned re-laparotomy and the need for optimization of physiology and immunology. Eur J Trauma Emerg Surg 2014; 40:135-42. [PMID: 26815893 DOI: 10.1007/s00068-014-0396-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Accepted: 03/10/2014] [Indexed: 12/31/2022]
Abstract
Planned re-laparotomy or damage control laparotomy (DCL), first described by Dr. Harlan Stone in 1983, has become a widely utilized technique in a broad range of patients and operative situations. Studies have validated the use of DCL by demonstrating decreased mortality and morbidity in trauma, general surgery and abdominal vascular catastrophes. Indications for planned re-laparotomy include severe physiologic derangements, coagulopathy, concern for bowel ischemia, and abdominal compartment syndrome. The immunology of DCL patients is not well described in humans, but promising animal studies suggest a benefit from the open abdomen (OA) and several human trials on this subject are currently underway. Optimal critical care of patients with OA's, including sedation, paralysis, nutrition, antimicrobial and fluid management strategies have been associated with improved closure rates and recovery.
Collapse
Affiliation(s)
- L Kobayashi
- Division of Trauma, Surgical Critical Care, and Burns, Department of Surgery, University of California, San Diego, 200 W. Arbor Dr. #8896, San Diego, CA, 92103-8896, USA.
| | - R Coimbra
- Division of Trauma, Surgical Critical Care, and Burns, Department of Surgery, University of California, San Diego, 200 W. Arbor Dr. #8896, San Diego, CA, 92103-8896, USA.
| |
Collapse
|
145
|
Abstract
BACKGROUND Parenteral nutrition (PN) increases risks of infections in critically injured patients. Recently, PN was shown to reduce intestine luminal levels of the Paneth cell antimicrobial molecule secretory phospholipase A2 (sPLA2) and the goblet cell glycoprotein mucin2 (MUC2). These molecules are critical factors for innate mucosal immunity and provide barrier protection. Interleukin-4 (IL-4) and IL-13 regulate sPLA2 and MUC2 production through the IL-13 receptor. Because IL-25 stimulates IL-4 and IL-13 release and PN reduces luminal sPLA2 and MUC2, we hypothesized that adding IL-25 to PN would restore these innate immune factors and maintain barrier function. METHODS Two days after venous cannulation, male ICR (Institute of Cancer Research) mice were randomized to receive chow (n = 12), PN (n = 9), or PN + 0.7 μg of exogenous IL-25 (n = 11) daily for 5 days. Small-intestine wash fluid (SIWF) was collected for analysis of sPLA2 activity, MUC2 density, and luminal levels of IL-4 and IL-13. Small-intestinal tissue was harvested for analysis of tissue sPLA2 activity or immediate use in an ex-vivo intestinal segment culture (EVISC) to assess susceptibility of the tissue segments to enteroinvasive Escherichia coli. RESULTS PN reduced luminal sPLA2 (P < 0.0001) and MUC2 (P <0.002) compared with chow, whereas the addition of IL-25 to PN increased luminal sPLA2 (P < 0.0001) and MUC2 (P < 0.02) compared with PN. Tissue IL-4 and IL-13 decreased with PN compared with chow (IL-4: P < 0.0001, IL-13: P < 0.002), whereas IL-25 increased both cytokines compared with PN (IL-4: P < 0.03, IL-13: P < 0.02). Tissue levels of sPLA2 were significantly decreased with PN compared with chow, whereas IL-25 significantly increased tissue sPLA2 levels compared with PN alone. Functionally, more bacteria invaded the PN-treated tissue compared with chow (P < 0.01), and the addition of IL-25 to PN decreased enteroinvasion to chow levels (P < 0.01). CONCLUSIONS PN impairs innate mucosal immunity by suppressing luminal sPLA2 activity and MUC2 density compared with chow. PN also increases bacterial invasion in ex-vivo tissue. Administration of exogenous IL-25 reverses this dysfunction and increases luminal sPLA2 and MUC2. PN tissue treated with IL-25 was significantly more resistant to bacterial invasion than with PN alone, suggesting that IL-25-induced effects augment the barrier defense mechanisms.
Collapse
|
146
|
Lefrant JY, Hurel D, Cano NJ, Ichai C, Preiser JC, Tamion F. [Guidelines for nutrition support in critically ill patient]. ACTA ACUST UNITED AC 2014; 33:202-18. [PMID: 24565944 DOI: 10.1016/j.annfar.2014.01.008] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- J-Y Lefrant
- Services des réanimations, division anesthésie réanimation douleur urgence, CHU de Nîmes, place du Pr-Robert-Debré, 30029 Nîmes cedex 9, France.
| | - D Hurel
- Service de réanimation médico-chirurgicale, centre hospitalier François-Quesnay, 2, boulevard Sully, 78201 Mantes-la-Jolie cedex, France
| | - N J Cano
- Service de nutrition, CHU de Clermont-Ferrand, 63003 Clermont-Ferrand cedex, France; Unité de nutrition humaine, Clermont université, université d'Auvergne, BP 10448, 63000 Clermont-Ferrand, France; Inra, UMR 1019, UNH, CRNH Auvergne, 63000 Clermont-Ferrand, France
| | - C Ichai
- Service de réanimation médico-chirurgicale, hôpital Saint-Roch, CHU de Nice, 5, rue Pierre-Dévoluy, 06006 Nice cedex 1, France
| | - J-C Preiser
- Service des soins intensifs, hôpital universitaire Erasme, 808, route de Lennik, 1070 Bruxelles, Belgique
| | - F Tamion
- Service de réanimation médicale, hôpital Charles-Nicolle, CHU de Rouen, 1, rue de Germont, 76081 Rouen cedex, France
| |
Collapse
|
147
|
Wade CE, Kozar RA, Dyer CB, Bulger EM, Mourtzakis M, Heyland DK. Evaluation of nutrition deficits in adult and elderly trauma patients. JPEN J Parenter Enteral Nutr 2014; 39:449-55. [PMID: 24562001 DOI: 10.1177/0148607114523450] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Accepted: 01/17/2014] [Indexed: 12/13/2022]
Abstract
BACKGROUND As metabolism is often escalated following injury, severely injured trauma patients are at risk for underfeeding and adverse outcomes. METHODS From an international database of 12,573 critically ill, adult mechanically ventilated patients, who received a minimum of 3 days of nutrition therapy, trauma patients were identified and nutrition practices and outcomes compared with nontrauma patients. Within the trauma population, we compared nutrition practices and outcomes of younger vs older patients. RESULTS There were 1279 (10.2%) trauma patients. They were younger, were predominantly male, had lower Acute Physiology and Chronic Health Evaluation II (APACHE II) scores, and had an overall lower body mass index compared with nontrauma patients. Eighty percent of trauma patients received enteral feeding compared with 78% of nontrauma patients. Trauma patients were prescribed more calories and protein yet received similar amounts as nontrauma patients. Nutrition adequacy was reduced in both trauma and nontrauma patients. Survival was higher in trauma patients (86.6%) compared with nontrauma patients (71.8%). When patients who died were included as never discharged, trauma patients were more rapidly discharged from the intensive care unit (ICU) and hospital. Within the trauma population, 17.5% were elderly (≥65 years). The elderly had increased days of ventilation, ICU stay, and mortality compared with younger trauma patients. In a multivariable model, age and APACHE II score, but not nutrition adequacy, were associated with time to discharge alive from the hospital. CONCLUSION Significant nutrition deficits were noted in all patients. Elderly trauma patients have worse outcomes compared with younger patients. Further studies are necessary to evaluate whether increased nutrition intake can improve the outcomes of trauma patients, especially geriatric trauma patients.
Collapse
Affiliation(s)
- Charles E Wade
- The University of Texas Health Science Center at Houston, Houston
| | - Rosemary A Kozar
- The University of Texas Health Science Center at Houston, Houston
| | - Carmel B Dyer
- The University of Texas Health Science Center at Houston, Houston
| | | | | | | |
Collapse
|
148
|
Abstract
We published one of the first prospective randomized controlled trials evaluating early postoperative tube feeding (TF) in liver transplant recipients nearly 20 years ago. That first study showed that early posttransplant TF was safe and well tolerated; the study results also suggested that early TF could reduce posttransplant infection rates. This Pivotal Paper review evaluates the past, present, and future of early postoperative TF in liver transplantation. This article identifies what nutrition support findings more than 2 decades ago were the basis for attempting postoperative TF in liver transplantation. The results of our study, its unique findings, and shortcomings are summarized. Other subsequent studies of post-liver transplant TF are evaluated with a focus on effects on posttransplant infection rates. Finally, current transplant challenges, including donor organ shortage, increased severity of patients' pretransplant condition, expansion of living donor options, changes in immunosuppression, and use of specialized nutrients, are discussed in the context of how and why these factors affect nutrition support.
Collapse
Affiliation(s)
- Jeanette M Hasse
- Jeanette M. Hasse, CNSC, Annette C. and Harold C. Simmons Transplant Institute, Baylor University Medical Center, 3410 Worth St, Suite 950, Dallas, TX 75246, USA.
| |
Collapse
|
149
|
An integrated systematic review and meta-analysis of published randomized controlled trials evaluating nasogastric against postpyloris (nasoduodenal and nasojejunal) feeding in critically ill patients admitted in intensive care unit. Eur J Clin Nutr 2014; 68:424-32. [DOI: 10.1038/ejcn.2014.6] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Revised: 07/23/2013] [Accepted: 09/11/2013] [Indexed: 02/06/2023]
|
150
|
Mikhailov TA, Kuhn EM, Manzi J, Christensen M, Collins M, Brown AM, Dechert R, Scanlon MC, Wakeham MK, Goday PS. Early enteral nutrition is associated with lower mortality in critically ill children. JPEN J Parenter Enteral Nutr 2014; 38:459-66. [PMID: 24403379 DOI: 10.1177/0148607113517903] [Citation(s) in RCA: 105] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
BACKGROUND The purpose of this study was to examine the association of early enteral nutrition (EEN), defined as the provision of 25% of goal calories enterally over the first 48 hours of admission, with mortality and morbidity in critically ill children. METHODS We conducted a multicenter retrospective study of patients in 12 pediatric intensive care units (PICUs). We included patients aged 1 month to 18 years who had a PICU length of stay (LOS) of ≥96 hours for the years 2007-2008. We obtained patients' demographics, weight, Pediatric Index of Mortality-2 (PIM2) score, LOS, duration of mechanical ventilation (MV), mortality data, and nutrition intake data in the first 4 days after admission. RESULTS We identified 5105 patients (53.8% male; median age, 2.4 years). Mortality was 5.3%. EEN was achieved by 27.1% of patients. Children receiving EEN were less likely to die than those who did not (odds ratio, 0.51; 95% confidence interval, 0.34-0.76; P = .001 [adjusted for propensity score, PIM2 score, age, and center]). Comparing those who received EEN to those who did not, adjusted for PIM2 score, age, and center, LOS did not differ (P = .59), and the duration of MV for those receiving EEN tended to be longer than for those who did not, but the difference was not significant (P = .058). CONCLUSIONS EEN is strongly associated with lower mortality in patients with PICU LOS of ≥96 hours. LOS and duration of MV are slightly longer in patients receiving EEN, but the differences are not statistically significant.
Collapse
|