Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Sheila K. Jacobi is active.

Publication


Featured researches published by Sheila K. Jacobi.


Journal of Nutrition | 2012

Fish Oil Enhances Intestinal Integrity and Inhibits TLR4 and NOD2 Signaling Pathways in Weaned Pigs after LPS Challenge

Yulan Liu; Feng Chen; Jack Odle; Xi Lin; Sheila K. Jacobi; Huiling Zhu; Zhifeng Wu; Yongqing Hou

Long-chain (n-3) PUFA exert beneficial effects on inflammatory bowel diseases in animal models and clinical trials. In addition, pattern recognition receptors such as toll-like receptors (TLR) and nucleotide-binding oligomerization domain proteins (NOD) play a critical role in intestinal inflammation. We hypothesized that fish oil could alleviate Escherichia coli LPS-induced intestinal injury via modulation of TLR4 and NOD signaling pathways. Twenty-four weaned piglets were used in a 2 × 2 factorial design and the main factors included a dietary treatment (5% corn oil or 5% fish oil) and immunological challenge (LPS or saline). After feeding fish oil or corn oil diets for 21 d, pigs were injected with LPS or saline. At 4 h postinjection, blood samples were collected and pigs were killed. EPA, DHA, and total (n-3) PUFA were enriched in intestinal mucosa through fish supplementation. Fish oil improved intestinal morphology, indicated by greater villus height and villus height:crypt depth ratio, and intestinal barrier function, indicated by decreased plasma diamine oxidase (DAO) activity and increased mucosal DAO activity as well as enhanced protein expression of intestinal tight junction proteins including occludin and claudin-1. Moreover, fish oil decreased intestinal TNFα and PGE(2) concentrations and caspase-3 and heat shock protein 70 protein expression. Finally, fish oil downregulated the mRNA expression of intestinal TLR4 and its downstream signals myeloid differentiation factor 88, IL-1 receptor-associated kinase 1, TNFα receptor-associated factor 6, and NOD2, and its adaptor molecule, receptor-interacting serine/threonine-protein kinase 2. Fish oil decreased the protein expression of intestinal NFκB p65. These results indicate that fish oil supplementation is associated with inhibition of TLR4 and NOD2 signaling pathways and concomitant improvement of intestinal integrity under an inflammatory condition.


Advances in Nutrition | 2012

Nutritional Factors Influencing Intestinal Health of the Neonate

Sheila K. Jacobi; Jack Odle

Dietary nutrients are essential for gastrointestinal (GI) growth and function, and nutritional support of GI growth and development is a significant component of infant care. For healthy full-term neonates, nutritional provisions of the mothers milk and/or formula will support normal maturation of structure and function of the GI tract in most infants. The composition of breast milk affects GI barrier function and development of a competent mucosal immune system. The functional nutrients and other bioactive components of milk support a microenvironment for gut protection and maturation. However, premature infants struggle with feeding tolerance impairing normal GI function, leading to intestinal dysfunction and even death. The high prevalence worldwide of enteric diseases and dysfunction in neonates has led to much interest in understanding the role of nutrients and food components in the establishment and maintenance of a functioning GI tract. Neonates who do not receive enteral feeding as either mothers milk or formula are supported by total parental nutrition (TPN). The lack of enteral nutrition can compound intestinal dysfunction, leading to high morbidity and mortality in intestinally compromised infants. Reciprocally, enteral stimulation of an immature GI tract can also compound intestinal dysfunction. Therefore, further understanding of nutrient interactions with the mucosa is necessary to define nutritional requirements of the developing GI tract to minimize intestinal complications and infant morbidity. Piglet models of intestinal development and function are similar to humans, and this review summarizes recent findings regarding nutrient requirements for growth and maintenance of intestinal health. In particular, this article reviews the role of specific amino acids (arginine, glutamine, glutamate, and threonine), fatty acids (long chain polyunsaturated, medium chain, and short chain), various prebiotic carbohydrates (short-chain fructo-oligosaccharide, fructo--oligosaccharide, lacto-N-neotetraose, human milk oligosaccharide, polydextrose, and galacto-oligosaccharide), and probiotics that have been examined in the suckling piglet model of intestinal health.


Gut microbes | 2011

Acute necrotizing enterocolitis of preterm piglets is characterized by dysbiosis of ileal mucosa-associated bacteria

M. Andrea Azcarate-Peril; Derek M. Foster; Maria Belen Cadenas; Maria R. Stone; Sheila K. Jacobi; Stephen H. Stauffer; Anthony Pease; Jody L. Gookin

Investigation of bacteria involved in pathogenesis of necrotizing enterocolitis (NEC) is limited by infant fragility, analysis restricted to feces, use of culture-based methods, and lack of clinically-relevant animal models. This study used a unique preterm piglet model to characterize spontaneous differences in microbiome composition of NEC-predisposed regions of gut. Preterm piglets (n=23) were cesarean-delivered and nurtured for 30 hours over which time 52% developed NEC. Bacterial DNA from ileal content, ileal mucosa, and colonic mucosa were PCR amplified, subjected to terminal restriction fragment length polymorphism (TRFLP) analysis and targeted 16S rDNA qPCR. Preterm ileal mucosa was specifically bereft in diversity of bacteria compared to ileal content and colonic mucosa. Preterm ileum was restricted to representation by only Proteobacteria, Firmicutes, Cyanobacteria and Chloroflexi. In piglets with NEC, ileal mucosa was uniquely characterized by increases in number of Firmicutes and diversity of phyla to include Actinobacteria and uncultured bacteria. Five specific TRFLP profiles, corresponding in closest identity to Clostridium butyricum, C. neonatale, C. proteolyticum, Streptomyces spp., and Leptolyngbya spp., were significantly more prevalent or observed only among samples from piglets with NEC. Total numbers of Clostridium spp. and C. butyricum were significantly greater in samples of NEC ileal mucosa but not ileal content or colonic mucosa. These results provide strong support for ileal mucosa as a focus for investigation of specific dysbiosis associated with NEC and suggest a significant role for Clostridium spp., and members of the Actinobacteria and Cyanobacteria in the pathogenesis of NEC in preterm piglets.


Journal of Nutrition | 2011

Polydextrose Enrichment of Infant Formula Demonstrates Prebiotic Characteristics by Altering Intestinal Microbiota, Organic Acid Concentrations, and Cytokine Expression in Suckling Piglets

Tina M. Herfel; Sheila K. Jacobi; Xi Lin; Vivek Fellner; D. Carey Walker; Zeina Jouni; Jack Odle

Oligosaccharides, the 3rd-most abundant component in human milk, are virtually absent from infant formulas and from the cow milk on which most are based. In breast-fed infants, human milk oligosaccharides (HMO) act as both receptor analogs, interfering with pathogen adhesion, and as prebiotics, stimulating the growth of certain commensal bacteria (e.g. bifidobacteria) and supporting the innate immunity. To further align the functional properties of infant formula with those of human milk, polydextrose (PDX) is proposed as a substitute for HMO. To determine the prebiotic functionality of PDX, 1-d-old pigs were fed a cow milk-based formula supplemented with increasing concentrations of PDX (0, 1.7, 4.3, 8.5, or 17 g/L) for 18 d (n = 13). Additional reference groups included pigs sampled at d 0 and sow-reared pigs sampled at d 18 (n = 12). Ileal Lactobacilli CFU, but not Bifidobacteria, increased linearly with increasing PDX (P = 0.02). The propionic acid concentration in digesta linearly increased with the PDX level (P = 0.045) and lactic acid increased linearly by 5-fold with increasing PDX (P = 0.001). Accordingly, digesta pH decreased linearly (P < 0.05) as PDX increased, with a maximal reduction approaching 0.5 pH units in pigs fed 17 g/L. Expression of TNFα, IL-1β, and IL-8 showed a negative quadratic pattern in response to PDX supplementation, declining at intermediate concentrations and rebounding at higher concentrations of PDX. In summary, PDX enrichment of infant formula resulted in a prebiotic effect by increasing ileal lactobacilli and propionic and lactic acid concentrations and decreasing pH with associated alterations in ileal cytokine expression.


Journal of Nutrition | 2012

Dietary Long-Chain PUFA Enhance Acute Repair of Ischemia-Injured Intestine of Suckling Pigs

Sheila K. Jacobi; Adam J. Moeser; Benjamin A. Corl; Robert J. Harrell; Jack Odle

Abstract Infant formula companies have been fortifying formulas with long-chain PUFA for 10 y. Long-chain PUFA are precursors of prostanoids, which stimulate recovery of intestinal barrier function. Supplementation of milk with PUFA increases the content of arachidonic acid (ARA) in enterocyte membranes; however, the effect of this enrichment on intestinal repair is not known. The objective of these experiments was to investigate the effect of supplemental ARA on intestinal barrier repair in ischemia-injured porcine ileum. One-day-old pigs (n = 24) were fed a milk-based formula for 10 d. Diets contained no PUFA (0% ARA), 0.5% ARA, 5% ARA, or 5% EPA of total fatty acids. Following dietary enrichment, ilea were subjected to in vivo ischemic injury by clamping the local mesenteric blood supply for 45 min. Following the ischemic period, control (nonischemic) and ischemic loops were mounted on Ussing chambers. Transepithelial electrical resistance (TER) was measured over a 240-min recovery period. Ischemia-injured ileum from piglets fed 5% ARA (61.0 ± 14%) exhibited enhanced recovery compared with 0% ARA (16 ± 14) and 0.5% ARA (22.1 ± 14)-fed pigs. Additionally, ischemia-injured ileum from 5% EPA (51.3 ± 14)-fed pigs had enhanced recovery compared with 0% ARA-fed pigs (P < 0.05). The enhanced TER recovery response observed with ischemia-injured 5% ARA supplementation was supported by a significant reduction in mucosal-to-serosal flux of3H-mannitol and14C-inulin compared with all other ischemia-injured dietary groups (P < 0.05). A histological evaluation of ischemic ilea from piglets fed the 5% ARA showed reduced histological lesions after ischemia compared with the other dietary groups (P < 0.05). These data demonstrate that feeding elevated levels of long-chain PUFA enhances acute recovery of ischemia-injured porcine ileum.


Journal of Nutrition | 2011

Dietary Arachidonate Differentially Alters Desaturase-Elongase Pathway Flux and Gene Expression in Liver and Intestine of Suckling Pigs

Sheila K. Jacobi; Xi Lin; Benjamin A. Corl; Holly A. Hess; Robert J. Harrell; Jack Odle

Because dietary arachidonate (ARA) and its eicosanoid derivatives are major regulators of intestinal homeostasis and repair following injury, we evaluated the effects of dietary ARA on desaturation and elongation of (13)C-18:2(n-6) and mRNA abundance of Δ-6-desaturase (FADS2), elongase (ELOVL5), and Δ-5-desaturase (FADS1) in liver and intestine. Day-old pigs (n = 96) were fed milk-based formula containing 0, 0.5, 2.5, or 5% ARA or 5% eicosapentaenoic acid of total fatty acids for 4, 8, and 16 d. In liver, the desaturation rate [nmol/(g tissue⋅h)] of (13)C-18:2(n-6) to (13)C-18:3(n-6) decreased 56% between 4 and 16 d but was not affected by diet. Whereas accumulation in (13)C-20:3(n-6) also decreased with age by 67%, it increased linearly with increasing dietary ARA (P < 0.06). In comparison, intestinal flux was ~50% less than liver flux and was unaffected by age, but desaturation to (13)C-18:3(n-6) increased linearly (by 57%) in pigs fed ARA diets (P < 0.001), equaling the rate observed in sow-fed controls. In both liver and intestine, alternate elongation to (13)C-20:2(n-6) (via Δ-8-desaturase) was markedly elevated in pigs fed the 0% ARA diet compared with all other dietary treatments (P < 0.01). Transcript abundance of FADS2, ELOVL5, and FADS1 was not affected in liver by diet (P > 0.05) but decreased precipitously between birth and d 4 (~70%; P < 0.05). In contrast, intestinal abundance of FADS2 and FADS1 increased 60% from d 4 to 16. In conclusion, dietary ARA regulated the desaturase-elongase pathway in a tissue-specific manner. In liver, ARA had modest effects on (n-6) fatty acid flux, and intestinal FADS2 activity and mRNA increased. Additionally, hepatic flux decreased with postnatal age, whereas intestinal flux did not change.


Journal of Nutrition | 2008

Enrichment of Intestinal Mucosal Phospholipids with Arachidonic and Eicosapentaenoic Acids Fed to Suckling Piglets Is Dose and Time Dependent

Holly A. Hess; Benjamin A. Corl; Xi Lin; Sheila K. Jacobi; Robert J. Harrell; Jack Odle

Infant formula companies began fortifying formulas with long-chain PUFA in 2002, including arachidonic acid (ARA) at approximately 0.5% of total fatty acids. The primary objective of this study was to determine the time-specific effects of feeding formula enriched with supra-physiologic ARA on fatty acid composition of intestinal mucosal phospholipids. One-day-old pigs (n = 96) were fed a milk-based formula for 4, 8, or 16 d. Diets contained either no PUFA (0% ARA, negative control), 0.5% ARA, 2.5% ARA, 5% ARA, or 5% eicosapentaenoic acid (EPA) of total fatty acids (wt:wt). Growth (299 +/- 21 g/d) and clinical hematology were unaffected by treatment (P > 0.6). Although minimal on d 4, concentrations of ARA in jejunal mucosa were enriched 47, 272 and 428% by d 8 and 144, 356, and 415% by d 16 in pigs fed the 0.5% ARA, 2.5% ARA, and 5% ARA diets, respectively, compared with the 0% ARA control pigs (P < 0.01). On d 16, ARA enrichment increased progressively with increasing dietary ARA supplementation from 0 to 2.5% but plateaued as dietary ARA rose to 5%. A similar pattern of ARA enrichment was observed in ileal mucosal phospholipids, but maximal enrichment in the ileum exceed that in the jejunum by >50%. As ARA increased, linoleic acid content decreased reciprocally. Although maximal enterocyte enrichment with EPA approached 20-fold by d 8, concentrations were only approximately 50% of those attained for ARA. Negligible effects on gross villus/crypt morphology were observed. These data demonstrate a dose-dependent response of intestinal mucosal phospholipid ARA concentration to dietary ARA with nearly full enrichment attained within 8 d of feeding formula containing ARA at 2.5% of total fatty acids and that supra-physiologic supplementation of ARA is not detrimental to growth.


Food and Chemical Toxicology | 2009

Safety evaluation of polydextrose in infant formula using a suckling piglet model.

Tina M. Herfel; Sheila K. Jacobi; Xi Lin; D.C. Walker; Z.E. Jouni; Jack Odle

Oligosaccharides, the third largest component in human milk, are virtually absent from cows milk and most infant formula. Prebiotic carbohydrates like polydextrose (PDX) have been proposed as surrogates for human milk oligosaccharides. Safety assessments of novel infant formula ingredients include dose-response experiments in appropriate neonatal animal models such as the suckling pig. To further substantiate the safety of the ingredient, one-day old pigs were fed a cows milk-based formula supplemented with PDX (1.7, 4.3, 8.5 or 17 g/L) for 18 days (n=13/dose) and compared to appropriate control (unsupplemented formula; n=13) and reference groups (day 0 pigs, and sow-reared pigs; n=13). Growth rate, formula intake, stool consistency, behavior score, blood chemistry and hematology, relative organ weights (% of body weight), tissue morphology (i.e. liver, kidney and pancreas) and pancreas biochemistry did not differ among formula-fed pigs (P>0.1). Polydextrose mimicked other prebiotic carbohydrates and had no adverse effect at the highest tested level 17.0 g PDX/L, equivalent to a dose of 8.35 g/kg of body weight per day.


World Journal of Gastroenterology | 2013

Acute effects of rotavirus and malnutrition on intestinal barrier function in neonatal piglets

Sheila K. Jacobi; Adam J. Moeser; J. Marc Rhoads; Benjamin A. Corl; Robert J. Harrell; Jack Odle

AIM To investigate the effect of protein-energy malnutrition on intestinal barrier function during rotavirus enteritis in a piglet model. METHODS Newborn piglets were allotted at day 4 of age to the following treatments: (1) full-strength formula (FSF)/noninfected; (2) FSF/rotavirus infected; (3) half-strength formula (HSF)/noninfected; or (4) HSF/rotavirus infected. After one day of adjustment to the feeding rates, pigs were infected with rotavirus and acute effects on growth and diarrhea were monitored for 3 d and jejunal samples were collected for Ussing-chamber analyses. RESULTS Piglets that were malnourished or infected had lower body weights on days 2 and 3 post-infection (P < 0.05). Three days post-infection, marked diarrhea and weight loss were accompanied by sharp reductions in villus height (59%) and lactase activity (91%) and increased crypt depth (21%) in infected compared with non-infected pigs (P < 0.05). Malnutrition also increased crypt depth (21%) compared to full-fed piglets. Villus:crypt ratio was reduced (67%) with viral infection. There was a trend for reduction in transepithelial electrical resistance with rotavirus infection and malnutrition (P = 0.1). (3)H-mannitol flux was significantly increased (50%; P < 0.001) in rotavirus-infected piglets compared to non-infected piglets, but there was no effect of nutritional status. Furthermore, rotavirus infection reduced localization of the tight junction protein, occludin, in the cell membrane and increased localization in the cytosol. CONCLUSION Overall, malnutrition had no additive effects to rotavirus infection on intestinal barrier function at day 3 post-infection in a neonatal piglet model.


Journal of Nutritional Biochemistry | 2011

Dietary conjugated linoleic acid alters long chain polyunsaturated fatty acid metabolism in brain and liver of neonatal pigs

Xi Lin; Jenny Bo; Susan A. Mathews Oliver; Benjamin A. Corl; Sheila K. Jacobi; William T. Oliver; Robert J. Harrell; Jack Odle

Effects of dietary conjugated linoleic acid (CLA, 1% mixed isomers) on n-6 long-chain polyunsaturated fatty acid (LCPUFA) oxidation and biosynthesis were investigated in liver and brain tissues of neonatal piglets. Fatty acid β-oxidation was measured in tissue homogenates using [1-(14)C]linoleic acid (LA) and -arachidonic acid (ARA) substrates, while fatty acid desaturation and elongation were traced using [U-(13)C]LA and GC-MS. Dietary CLA had no effect on fatty acid β-oxidation, but significantly decreased n-6 LCPUFA biosynthesis by inhibition of LA elongation and desaturation. Differences were noted between our (13)C tracer assessment of desaturation/elongation and simple precursor-product indices computed from fatty acid composition data, indicating that caution should be exercised when employing the later. The inhibitory effects of CLA on elongation/desaturation were more pronounced in pigs fed a low fat diet (3% fat) than a high fat diet (25% fat). Direct elongation of linoleic acid to C20:2n-6 via the alternate elongation pathway might play an important role in n-6 LCPUFA synthesis because more than 40% of the synthetic products of [U-(13)C]LA accumulated in [(13)C]20:2n-6. Overall, the data show that dietary CLA shifted the distribution of the synthetic products of [U-(13)C]LA between elongation and desaturation in liver and decreased the total synthetic products of [U-(13)C]LA in brain by inhibiting LA elongation to C20:2n-6. The impact of CLA on brain LCPUFA metabolism of the developing neonate merits consideration and further investigation.

Collaboration


Dive into the Sheila K. Jacobi's collaboration.

Top Co-Authors

Avatar

Jack Odle

North Carolina State University

View shared research outputs
Top Co-Authors

Avatar

Xi Lin

North Carolina State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Robert J. Harrell

North Carolina State University

View shared research outputs
Top Co-Authors

Avatar

Tina M. Herfel

North Carolina State University

View shared research outputs
Top Co-Authors

Avatar

Chaolai Man

North Carolina State University

View shared research outputs
Top Co-Authors

Avatar

Holly A. Hess

North Carolina State University

View shared research outputs
Top Co-Authors

Avatar

Hope Lima

North Carolina State University

View shared research outputs
Top Co-Authors

Avatar

Lin Xi

North Carolina State University

View shared research outputs
Top Co-Authors

Avatar

Adam J. Moeser

Michigan State University

View shared research outputs
Researchain Logo
Decentralizing Knowledge