Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Front Immunol ; 12: 665968, 2021.
Article in English | MEDLINE | ID: mdl-34135894

ABSTRACT

Through food intake, humans obtain a variety of nutrients that are essential for growth, cellular function, tissue development, energy, and immune defense. A special interaction between nutrients and gut-associated lymphoid tissue occurs in the intestinal tract. Enterocytes of the intestinal barrier act as sensors for antigens from nutrients and the intestinal microbiota, which they deliver to the underlying immune system of the lamina propria, triggering an immune response. Studies investigating the mechanism of influence of nutrition on immunological outcomes have highlighted an important role of macronutrients (proteins, carbohydrates, fatty acids) and micronutrients (vitamins, minerals, phytochemicals, antioxidants, probiotics) in modulating immune homeostasis. Nutrients exert their role in innate immunity and inflammation by regulating the expression of TLRs, pro- and anti-inflammatory cytokines, thus interfering with immune cell crosstalk and signaling. Chemical substrates derived from nutrient metabolism may act as cofactors or blockers of enzymatic activity, influencing molecular pathways and chemical reactions associated with microbial killing, inflammation, and oxidative stress. Immune cell function appears to be influenced by certain nutrients that form parts of the cell membrane structure and are involved in energy production and prevention of cytotoxicity. Nutrients also contribute to the initiation and regulation of adaptive immune responses by modulating B and T lymphocyte differentiation, proliferation and activation, and antibody production. The purpose of this review is to present the available data from the field of nutritional immunology to elucidate the complex and dynamic relationship between nutrients and the immune system, the delineation of which will lead to optimized nutritional regimens for disease prevention and patient care.


Subject(s)
Immunity/immunology , Intestinal Mucosa/immunology , Lymphoid Tissue/immunology , Nutrients/immunology , Antibodies/immunology , Antigen-Presenting Cells/immunology , Cytokines/immunology , Gastrointestinal Microbiome/immunology , Humans , Intestinal Mucosa/microbiology , Lymphocytes/immunology , Micronutrients/deficiency , Micronutrients/immunology , Nutrients/deficiency
2.
Cancer Immunol Immunother ; 70(12): 3435-3449, 2021 Dec.
Article in English | MEDLINE | ID: mdl-33877384

ABSTRACT

Specific extracts of selected vegetables (SV) have been shown to benefit the survival of stage IIIb/IV non-small cell lung cancer patients in phase I/II studies and is currently in a phase III trial. However, the underlying mechanism of SV-mediated antitumor immune responses has not been elucidated. Our results indicate that SV modulated the NK and adoptive T cell immune responses in antitumor efficacy. Furthermore, antitumor effects of SV were also mediated by innate myeloid cell function, which requires both TLR and ß-glucan signaling in a MyD88/TRIF and Dectin-1-dependent manner, respectively. Additionally, SV treatment reduced granulocytic myeloid-derived suppressor cell (MDSC) infiltration into the tumor and limited monocytic MDSC toward the M2-like functional phenotype. Importantly, SV treatment enhanced antigen-specific immune responses by augmenting the activation of antigen-specific TH1/TH17 cells in secondary lymphoid organs and proliferative response, as well as by reducing the Treg population in the tumor microenvironment, which was driven by SV-primed activated M-MDSC. Our results support the idea that SV can subvert immune-tolerance state in the tumor microenvironment and inhibit tumor growth. The present study suggests that features, such as easy accessibility, favorable clinical efficacy, no detectable side effects and satisfactory safety make SV a feasible, appealing and convincing adjuvant therapy for the treatment of cancer patients and prevent tumor recurrence and/or metastases.


Subject(s)
Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/therapy , Lung Neoplasms/immunology , Lung Neoplasms/therapy , Nutrients/immunology , Plant Extracts/immunology , Tumor Microenvironment/immunology , Animals , Dietary Supplements , Disease Models, Animal , Immune Tolerance/immunology , Immunity/immunology , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Mice, Transgenic , Monocytes/immunology , Myeloid Cells/immunology , Myeloid-Derived Suppressor Cells/immunology , Neoplasm Recurrence, Local/immunology , Th1 Cells/immunology , Th17 Cells/immunology
3.
Nutrients ; 13(4)2021 Mar 24.
Article in English | MEDLINE | ID: mdl-33804909

ABSTRACT

Optimal nutrition is the foundation for the development and maintenance of a healthy immune system. An optimal supply of nutrients is required for biosynthesis of immune factors and immune cell proliferation. Nutrient deficiency/inadequacy and hidden hunger, which manifests as depleted nutrients reserves, increase the risk of infectious diseases and aggravate disease severity. Therefore, an adequate and balanced diet containing an abundant diversity of foods, nutrients, and non-nutrient chemicals is paramount for an optimal immune defense against infectious diseases, including cold/flu and non-communicable diseases. Some nutrients and foods play a larger role than others in the support of the immune system. Oats are a nutritious whole grain and contain several immunomodulating nutrients. In this narrative review, we discuss the contribution of oat nutrients, including dietary fiber (ß-glucans), copper, iron, selenium, and zinc, polyphenolics (ferulic acid and avenanthramides), and proteins (glutamine) in optimizing the innate and adaptive immune system's response to infections directly by modulating the innate and adaptive immunity and indirectly by eliciting changes in the gut microbiota and related metabolites.


Subject(s)
Avena/immunology , Diet/methods , Immunity/immunology , Nutrients/administration & dosage , Nutrients/immunology , Dietary Fiber/administration & dosage , Humans
4.
Int Arch Allergy Immunol ; 182(4): 324-338, 2021.
Article in English | MEDLINE | ID: mdl-33567446

ABSTRACT

In this article, we propose that differences in COVID-19 morbidity may be associated with transient receptor potential ankyrin 1 (TRPA1) and/or transient receptor potential vanilloid 1 (TRPV1) activation as well as desensitization. TRPA1 and TRPV1 induce inflammation and play a key role in the physiology of almost all organs. They may augment sensory or vagal nerve discharges to evoke pain and several symptoms of COVID-19, including cough, nasal obstruction, vomiting, diarrhea, and, at least partly, sudden and severe loss of smell and taste. TRPA1 can be activated by reactive oxygen species and may therefore be up-regulated in COVID-19. TRPA1 and TRPV1 channels can be activated by pungent compounds including many nuclear factor (erythroid-derived 2) (Nrf2)-interacting foods leading to channel desensitization. Interactions between Nrf2-associated nutrients and TRPA1/TRPV1 may be partly responsible for the severity of some of the COVID-19 symptoms. The regulation by Nrf2 of TRPA1/TRPV1 is still unclear, but suggested from very limited clinical evidence. In COVID-19, it is proposed that rapid desensitization of TRAP1/TRPV1 by some ingredients in foods could reduce symptom severity and provide new therapeutic strategies.


Subject(s)
COVID-19/diet therapy , COVID-19/immunology , NF-E2-Related Factor 2/immunology , Nutrients/immunology , SARS-CoV-2/immunology , TRPA1 Cation Channel/immunology , TRPV Cation Channels/immunology , Antioxidants/metabolism , Biomarkers/metabolism , Brassica , COVID-19/complications , COVID-19/diagnosis , COVID-19 Testing , Desensitization, Immunologic/methods , Down-Regulation , Humans , Oxidative Stress/immunology , SARS-CoV-2/pathogenicity , Severity of Illness Index , Up-Regulation
5.
J Infect Dis ; 223(12 Suppl 2): S194-S200, 2021 06 16.
Article in English | MEDLINE | ID: mdl-33326565

ABSTRACT

Clostridioides difficile is an urgent antimicrobial-resistant bacterium, causing mild to moderate and sometimes life-threatening disease. Commensal gut microbes are critical for providing colonization resistance against C difficile and can be leveraged as non-antibiotic alternative therapeutics for the prevention and treatment of C difficile infection.


Subject(s)
Clostridioides difficile/growth & development , Clostridium Infections/immunology , Animals , Anti-Bacterial Agents/immunology , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/therapeutic use , Antibiosis , Bile Acids and Salts/immunology , Bile Acids and Salts/metabolism , Clostridioides difficile/drug effects , Clostridioides difficile/metabolism , Clostridium Infections/drug therapy , Clostridium Infections/microbiology , Fatty Acids, Volatile/immunology , Fatty Acids, Volatile/metabolism , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/physiology , Humans , Nutrients/immunology , Nutrients/metabolism
6.
Nutrients ; 12(6)2020 May 27.
Article in English | MEDLINE | ID: mdl-32471251

ABSTRACT

The coronavirus-disease 2019 (COVID-19) was announced as a global pandemic by the World Health Organization. Challenges arise concerning how to optimally support the immune system in the general population, especially under self-confinement. An optimal immune response depends on an adequate diet and nutrition in order to keep infection at bay. For example, sufficient protein intake is crucial for optimal antibody production. Low micronutrient status, such as of vitamin A or zinc, has been associated with increased infection risk. Frequently, poor nutrient status is associated with inflammation and oxidative stress, which in turn can impact the immune system. Dietary constituents with especially high anti-inflammatory and antioxidant capacity include vitamin C, vitamin E, and phytochemicals such as carotenoids and polyphenols. Several of these can interact with transcription factors such as NF-kB and Nrf-2, related to anti-inflammatory and antioxidant effects, respectively. Vitamin D in particular may perturb viral cellular infection via interacting with cell entry receptors (angiotensin converting enzyme 2), ACE2. Dietary fiber, fermented by the gut microbiota into short-chain fatty acids, has also been shown to produce anti-inflammatory effects. In this review, we highlight the importance of an optimal status of relevant nutrients to effectively reduce inflammation and oxidative stress, thereby strengthening the immune system during the COVID-19 crisis.


Subject(s)
Coronavirus Infections , Diet , Immune System/immunology , Inflammation/immunology , Nutrients/immunology , Oxidative Stress/immunology , Pandemics , Pneumonia, Viral , Antioxidants , Betacoronavirus , COVID-19 , Coronavirus Infections/immunology , Humans , Inflammation/prevention & control , Nutritional Status/immunology , Pneumonia, Viral/immunology , SARS-CoV-2
7.
mSphere ; 5(2)2020 03 11.
Article in English | MEDLINE | ID: mdl-32161145

ABSTRACT

Clostridioides difficile is a spore-forming bacterium that causes severe colitis and is a major public health threat. During infection, C. difficile toxin production results in damage to the epithelium and a hyperinflammatory response. The immune response to CDI leads to robust neutrophil infiltration at the sight of infection and the deployment of numerous antimicrobials. One of the most abundant host immune factors associated with CDI is calprotectin, a metal-chelating protein with potent antimicrobial activity. Calprotectin is essential to the innate immune response to C. difficile and increasing levels of calprotectin correlate with disease severity in both adults and children with CDI. The fact that C. difficile persists in the presence of high levels of calprotectin suggests that this organism may deploy strategies to compete with this potent antimicrobial factor for essential nutrient metals during infection. In this report, we demonstrate that a putative zinc (Zn) transporter, ZupT, is employed by C. difficile to survive calprotectin-mediated metal limitation. ZupT is highly expressed in the presence of calprotectin and is required to protect C. difficile against calprotectin-dependent growth inhibition. When competing against wild-type C. difficile, zupT mutants show a defect in colonization and persistence in a murine model of infection. Together these data demonstrate that C. difficile utilizes a metal import system to combat nutritional immunity during CDI and suggest that strategies targeting nutrient acquisition in C. difficile may have therapeutic potential.IMPORTANCE During infection, pathogenic organisms must acquire essential transition metals from the host environment. Through the process of nutritional immunity, the host employs numerous strategies to restrict these key nutrients from invading pathogens. In this study, we describe a mechanism by which the important human pathogen Clostridioides difficile resists transition-metal limitation by the host. We report that C. difficile utilizes a zinc transporter, ZupT, to compete with the host protein calprotectin for nutrient zinc. Inactivation of this transporter in C. difficile renders this important pathogen sensitive to host-mediated metal restriction and confers a fitness disadvantage during infection. Our study demonstrates that targeting nutrient metal transport proteins in C. difficile is a potential avenue for therapeutic development.


Subject(s)
Bacterial Proteins/immunology , Clostridioides difficile/pathogenicity , Clostridium Infections/immunology , Membrane Transport Proteins/immunology , Zinc/metabolism , Animals , Bacterial Proteins/genetics , Carrier Proteins/genetics , Carrier Proteins/immunology , Clostridioides difficile/genetics , Disease Models, Animal , Host-Pathogen Interactions/immunology , Humans , Immunity, Innate , Leukocyte L1 Antigen Complex/metabolism , Membrane Transport Proteins/genetics , Mice , Nutrients/immunology
8.
J Exp Med ; 217(1)2020 01 06.
Article in English | MEDLINE | ID: mdl-31649036

ABSTRACT

Foxp3+ regulatory T (T reg) cells are pivotal regulators of immune tolerance, with T cell receptor (TCR)-driven activated T reg (aT reg) cells playing a central role; yet how TCR signaling propagates to control aT reg cell responses remains poorly understood. Here we show that TCR signaling induces expression of amino acid transporters, and renders amino acid-induced activation of mTORC1 in aT reg cells. T reg cell-specific ablation of the Rag family small GTPases RagA and RagB impairs amino acid-induced mTORC1 signaling, causing defective amino acid anabolism, reduced T reg cell proliferation, and a rampant autoimmune disorder similar in severity to that triggered by T reg cell-specific TCR deficiency. Notably, T reg cells in peripheral tissues, including tumors, are more sensitive to Rag GTPase-dependent nutrient sensing. Ablation of RagA alone impairs T reg cell accumulation in the tumor, resulting in enhanced antitumor immunity. Thus, nutrient mTORC1 signaling is an essential component of TCR-initiated T reg cell reprogramming, and Rag GTPase activities may be titrated to break tumor immune tolerance.


Subject(s)
Immune Tolerance/immunology , Mechanistic Target of Rapamycin Complex 1/immunology , Nutrients/immunology , Signal Transduction/immunology , T-Lymphocytes, Regulatory/immunology , Amino Acid Transport Systems/immunology , Animals , Female , Male , Mice , Mice, Inbred C57BL , Monomeric GTP-Binding Proteins/immunology , Receptors, Antigen, T-Cell/immunology , TOR Serine-Threonine Kinases/immunology
9.
Trends Biochem Sci ; 44(12): 1041-1056, 2019 12.
Article in English | MEDLINE | ID: mdl-31326221

ABSTRACT

Zinc is an essential cofactor required for life and, as such, mechanisms exist for its homeostatic maintenance in biological systems. Despite the evolutionary distance between vertebrates and microbial life, there are parallel mechanisms to balance the essentiality of zinc with its inherent toxicity. Vertebrates regulate zinc homeostasis through a complex network of metal transporters and buffering systems that respond to changes in nutritional zinc availability or inflammation. Fine-tuning of this network becomes crucial during infections, where host nutritional immunity attempts to limit zinc availability to pathogens. However, accumulating evidence demonstrates that pathogens have evolved mechanisms to subvert host-mediated zinc withholding, and these metal homeostasis systems are important for survival within the host. We discuss here the mechanisms of vertebrate and bacterial zinc homeostasis and mobilization, as well as recent developments in our understanding of microbial zinc acquisition.


Subject(s)
Host-Pathogen Interactions/physiology , Infections/immunology , Nutrients/immunology , Zinc/immunology , Animals , Humans
10.
Nat Rev Immunol ; 19(5): 324-335, 2019 05.
Article in English | MEDLINE | ID: mdl-30820043

ABSTRACT

At the centre of the therapeutic dilemma posed by cancer is the question of how to develop more effective treatments that discriminate between normal and cancerous tissues. Decades of research have shown us that universally applicable principles are rare, but two well-accepted concepts have emerged: first, that malignant transformation goes hand in hand with distinct changes in cellular metabolism; second, that the immune system is critical for tumour control and clearance. Unifying our understanding of tumour metabolism with immune cell function may prove to be a powerful approach in the development of more effective cancer therapies. Here, we explore how nutrient availability in the tumour microenvironment shapes immune responses and identify areas of intervention to modulate the metabolic constraints placed on immune cells in this setting.


Subject(s)
Immunity/immunology , Neoplasms/immunology , Neoplasms/metabolism , Animals , Humans , Immunotherapy/methods , Nutrients/immunology , Nutrients/metabolism , Tumor Microenvironment/immunology
11.
Sci Immunol ; 4(32)2019 02 08.
Article in English | MEDLINE | ID: mdl-30737355

ABSTRACT

T cell responses to symbionts in the intestine drive tolerance or inflammation depending on the genetic background of the host. These symbionts in the gut sense the available nutrients and adapt their metabolic programs to use these nutrients efficiently. Here, we ask whether diet can alter the expression of a bacterial antigen to modulate adaptive immune responses. We generated a CD4+ T cell hybridoma, BθOM, specific for Bacteroides thetaiotaomicron (B. theta). Adoptively transferred transgenic T cells expressing the BθOM TCR proliferated in the colon, colon-draining lymph node, and spleen in B. theta-colonized healthy mice and differentiated into regulatory T cells (Tregs) and effector T cells (Teffs). Depletion of B. theta-specific Tregs resulted in colitis, showing that a single protein expressed by B. theta can drive differentiation of Tregs that self-regulate Teffs to prevent disease. We found that BθOM T cells recognized a peptide derived from a single B. theta protein, BT4295, whose expression is regulated by nutrients, with glucose being a strong catabolite repressor. Mice fed a high-glucose diet had a greatly reduced activation of BθOM T cells in the colon. These studies establish that the immune response to specific bacterial antigens can be modified by changes in the diet by altering antigen expression in the microbe.


Subject(s)
Antigens, Bacterial/metabolism , Bacteroides thetaiotaomicron/immunology , Colon/immunology , Diet , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer/methods , Animals , Antigens, Bacterial/immunology , Bacterial Proteins/metabolism , Cell Differentiation/immunology , Colitis/immunology , Colitis/prevention & control , Culture Media , Escherichia coli/immunology , Glucose/metabolism , Hybridomas/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nutrients/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism
12.
Biomed Pharmacother ; 109: 1000-1007, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30551349

ABSTRACT

Chronic fatigue syndrome (CFS) is known as a multi-systemic and complex illness, which induces fatigue and long-term disability in educational, occupational, social, or personal activities. The diagnosis of this disease is difficult, due to lacking a proper and suited diagnostic laboratory test, besides to its multifaceted symptoms. Numerous factors, including environmental and immunological issues, and a large spectrum of CFS symptoms, have recently been reported. In this review, we focus on the nutritional intervention in CFS, discussing the many immunological, environmental, and nutritional aspects currently investigated about this disease. Changes in immunoglobulin levels, cytokine profiles and B- and T- cell phenotype and declined cytotoxicity of natural killer cells, are commonly reported features of immune dysregulation in CFS. Also, some nutrient deficiencies (vitamin C, vitamin B complex, sodium, magnesium, zinc, folic acid, l-carnitine, l-tryptophan, essential fatty acids, and coenzyme Q10) appear to be important in the severity and exacerbation of CFS symptoms. This review highlights a far-driven analysis of mineral and vitamin deficiencies among CFS patients.


Subject(s)
Dietary Supplements , Fatigue Syndrome, Chronic/diet therapy , Fatigue Syndrome, Chronic/immunology , Gastrointestinal Microbiome/immunology , Nutrients/administration & dosage , Nutrients/immunology , Cytokines/immunology , Fatigue Syndrome, Chronic/microbiology , Gastrointestinal Microbiome/drug effects , Humans , Oxidative Stress/drug effects , Oxidative Stress/physiology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
13.
Methods Mol Biol ; 1862: 187-216, 2019.
Article in English | MEDLINE | ID: mdl-30315469

ABSTRACT

Immune cell function is tightly regulated by cellular metabolism, which in turn is strongly linked to the nutrient availability in the microenvironment surrounding the cells. This link is critical for effector CD8+ T cells which, after activation, must migrate from nutrient-rich environments into nutrient-scarce regions such as the tumor microenvironment. Assessing how nutrient availability modulates the metabolism of effector CD8+ T cells is thus key for understanding how harsh environments may impair their proliferation and effector function. Here, we describe an approach to systematically study the impact of the nutrient microenvironment on the metabolism of effector CD8+ T cells, based on performing stable 13C isotope labeling measurements on in vitro-differentiated murine effector CD8+ T cells.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Metabolomics/methods , Nutrients/metabolism , Animals , CD8-Positive T-Lymphocytes/immunology , Carbon Isotopes/chemistry , Cell Differentiation/immunology , Cells, Cultured , Culture Media/analysis , Culture Media/chemistry , Culture Media/metabolism , Lymphocyte Activation/immunology , Mass Spectrometry/instrumentation , Mass Spectrometry/methods , Metabolomics/instrumentation , Mice , Mice, Inbred C57BL , Nutrients/chemistry , Nutrients/immunology , Primary Cell Culture
14.
Vet Parasitol ; 264: 52-57, 2018 Dec 15.
Article in English | MEDLINE | ID: mdl-30503092

ABSTRACT

Haemonchus contortus is an economic problem in sheep farms worldwide, mainly in the tropics and subtropics. A vaccine against haemonchosis, called Barbervax®, was evaluated in ewes under two nutritional status, naturally infected with gastrointestinal nematodes. Ewes were divided into four groups: Supplemented Diet - Vaccine; Supplemented Diet - No vaccine; Basal Diet - Vaccine and Basal Diet - No vaccine. Their lambs were divided in Vaccinated and No vaccine. Ewes were immunised six times starting about 1 month of pregnancy with the first three doses at 3 week intervals and the last three shots at 4 week intervals. Supplemented ewes had higher body weight, body score and packed cell volume compared with those fed a basal diet. Both groups of vaccinated ewes showed a similar response in circulating anti-vaccine antibodies but the vaccine had no discernible effect on either body weight, body score and packed cell volume. There was a marked group difference in the number of ewes that received precautionary treatments with anthelmintic. All 14 Basal Diet - No vaccine ewes required treatment. In contrast only 7 ewes, in the Supplemented Diet - Vaccine group required anthelmintic treatment. In the Basal Diet - Vaccine and in the Supplemented Diet - No Vaccine groups, 12 and 13 ewes needed anthelmintic treatment, respectively. Vaccinated lambs showed much higher antibody titres resulting in 80% less Haemonchus spp. egg counts comparing with no vaccine lambs. Taken together these results clearly suggest that in pregnant and lactating ewes a combined protective effect between vaccination and improved nutrition resulted in fewer precautionary anthelmintic treatments. Thus, it was possible to achieve a more sustainable level of control of the haemonchosis, less dependent on anthelmintic drugs.


Subject(s)
Dietary Supplements , Haemonchiasis/veterinary , Nutrients/immunology , Sheep Diseases/immunology , Sheep Diseases/prevention & control , Vaccines/immunology , Animals , Antibodies, Helminth/blood , Female , Haemonchiasis/immunology , Haemonchiasis/prevention & control , Haemonchus/immunology , Nutrients/administration & dosage , Pregnancy , Sheep , Tropical Climate , Vaccines/administration & dosage , Weight Gain/immunology
15.
Int J Med Microbiol ; 308(1): 215-227, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29150190

ABSTRACT

The mature phagosome of macrophages is a hostile environment for the vast majority of phagocytosed microbes. In addition to active destruction of the engulfed microbes by antimicrobial compounds, restriction of essential nutrients in the phagosomal compartment contributes to microbial growth inhibition and killing. However, some pathogenic microorganisms have not only developed various strategies to efficiently withstand or counteract antimicrobial activities, but also to acquire nutrients within macrophages for intracellular replication. Successful intracellular pathogens are able to utilize host-derived amino acids, carbohydrates and lipids as well as trace metals and vitamins during intracellular growth. This requires sophisticated strategies such as phagosome modification or escape, efficient nutrient transporters and metabolic adaptation. In this review, we discuss the metabolic adaptation of facultative intracellular bacteria and fungi to the intracellular lifestyle inside macrophages.


Subject(s)
Adaptation, Physiological , Bacteria/metabolism , Cytosol/microbiology , Fungi/metabolism , Macrophages/microbiology , Animals , Biological Transport , Cytosol/metabolism , Humans , Macrophages/metabolism , Nutrients/immunology , Nutrients/metabolism , Phagosomes/metabolism , Phagosomes/microbiology
16.
J Hum Lact ; 34(1): 120-129, 2018 Feb.
Article in English | MEDLINE | ID: mdl-28614672

ABSTRACT

BACKGROUND: When human milk is unavailable, banked milk is recommended for feeding premature infants. Milk banks use processes to eliminate pathogens; however, variability among methods exists. Research aim: The aim of this study was to compare the macronutrient (protein, carbohydrate, fat, energy), immune-protective protein, and human milk oligosaccharide (HMO) content of human milk from three independent milk banks that use pasteurization (Holder vs. vat techniques) or retort sterilization. METHODS: Randomly acquired human milk samples from three different milk banks ( n = 3 from each bank) were analyzed for macronutrient concentrations using a Fourier transform mid-infrared spectroscopy human milk analyzer. The concentrations of IgA, IgM, IgG, lactoferrin, lysozyme, α-lactalbumin, α antitrypsin, casein, and HMO were analyzed by mass spectrometry. RESULTS: The concentrations of protein and fat were significantly ( p < .05) less in the retort sterilized compared with the Holder and vat pasteurized samples, respectively. The concentrations of all immune-modulating proteins were significantly ( p < .05) less in the retort sterilized samples compared with vat and/or Holder pasteurized samples. The total HMO concentration and HMOs containing fucose, sialic acid, and nonfucosylated neutral sugars were significantly ( p < .05) less in retort sterilized compared with Holder pasteurized samples. CONCLUSION: Random milk samples that had undergone retort sterilization had significantly less immune-protective proteins and total and specific HMOs compared with samples that had undergone Holder and vat pasteurization. These data suggest that further analysis of the effect of retort sterilization on human milk components is needed prior to widespread adoption of this process.


Subject(s)
Milk, Human/chemistry , Nutrients/chemistry , Oligosaccharides/chemistry , Caseins/analysis , Humans , Immunoglobulin A/analysis , Immunoglobulin G/analysis , Immunoglobulin M/analysis , Lactalbumin/analysis , Lactoferrin/analysis , Mass Spectrometry/methods , Milk Banks/statistics & numerical data , Milk, Human/immunology , Muramidase/analysis , Nutrients/immunology , Oligosaccharides/immunology , Pasteurization/methods
SELECTION OF CITATIONS
SEARCH DETAIL