1
|
Kim BJ, Kuhfeld RF, Haas JL, Anaya YM, Martinez RR, Sah BNP, Breen B, Newsham K, Malinczak CA, Dallas DC. Digestive Profiles of Human Milk, Recombinant Human and Bovine Lactoferrin: Comparing the Retained Intact Protein and Peptide Release. Nutrients 2024; 16:2360. [PMID: 39064803 PMCID: PMC11280017 DOI: 10.3390/nu16142360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 07/17/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
Lactoferrin (LF) is a major component of human milk. LF supplementation (currently bovine) supports the immune system and helps maintain iron homeostasis in adults. No recombinant human lactoferrin (rhLF) is available for commercial food use. To determine the extent to which rhLF (Effera™) produced by Komagataella phaffii digests similarly to hmLF, a validated in vitro digestion protocol was carried out. Bovine LF (bLF) was used as an additional control, as it is approved for use in various food categories. This study compared the extent of intact protein retention and the profile of peptides released in hmLF, bLF and rhLF (each with low and high iron saturation) across simulated adult gastric and intestinal digestion using gel electrophoresis, ELISA and LC-MS. Intact LF retention across digestion was similar across LF types, but the highest iron-saturated hmLF had greater retention in the simulated gastric fluid than all other sample types. Peptides identified in digested hmLF samples strongly correlated with digested rhLF samples (0.86 < r < 0.92 in the gastric phase and 0.63 < r < 0.70 in the intestinal phase), whereas digested bLF samples were significantly different. These findings support the potential for rhLF as a food ingredient for human consumption.
Collapse
Affiliation(s)
- Bum Jin Kim
- Nutrition Program, School of Nutrition and Public Health, College of Health, Oregon State University, Corvallis, OR 97331, USA; (B.J.K.); (R.F.K.); (B.N.P.S.)
| | - Russell F. Kuhfeld
- Nutrition Program, School of Nutrition and Public Health, College of Health, Oregon State University, Corvallis, OR 97331, USA; (B.J.K.); (R.F.K.); (B.N.P.S.)
| | - Joanna L. Haas
- Department of Food Science and Technology, Oregon State University, Corvallis, OR 97331, USA;
| | - Yanisa M. Anaya
- Helaina, New York, NY 10010, USA; (Y.M.A.); (R.R.M.); (B.B.); (K.N.); (C.-A.M.)
| | | | - Baidya Nath P. Sah
- Nutrition Program, School of Nutrition and Public Health, College of Health, Oregon State University, Corvallis, OR 97331, USA; (B.J.K.); (R.F.K.); (B.N.P.S.)
| | - Bella Breen
- Helaina, New York, NY 10010, USA; (Y.M.A.); (R.R.M.); (B.B.); (K.N.); (C.-A.M.)
| | - Kahler Newsham
- Helaina, New York, NY 10010, USA; (Y.M.A.); (R.R.M.); (B.B.); (K.N.); (C.-A.M.)
| | | | - David C. Dallas
- Nutrition Program, School of Nutrition and Public Health, College of Health, Oregon State University, Corvallis, OR 97331, USA; (B.J.K.); (R.F.K.); (B.N.P.S.)
- Department of Food Science and Technology, Oregon State University, Corvallis, OR 97331, USA;
| |
Collapse
|
2
|
Lu X, Cummings C, Osuala UA, Yennawar NH, Namitz KEW, Hellner B, Besada-Lombana PB, Peterson RD, Clark AJ. Characterization of recombinant human lactoferrin expressed in Komagataella phaffii. Analyst 2024; 149:3636-3650. [PMID: 38814097 PMCID: PMC11215759 DOI: 10.1039/d4an00333k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2024]
Abstract
This work presents a thorough characterization of Helaina recombinant human lactoferrin (rhLF, Effera™) expressed in a yeast system at an industrial scale for the first time. Proteomic analysis confirmed that its amino acid sequence is identical to that of native human LF. N-linked glycans were detected at three known glycosylation sites, namely, Asparagines-156, -497, and -642 and they were predominantly oligomannose structures having five to nine mannoses. Helaina rhLF's protein secondary structure was nearly identical to that of human milk lactoferrin (hmLF), as revealed by microfluidic modulation spectroscopy. Results of small-angle X-ray scattering (SAXS) and analytical ultracentrifugation analyses confirmed that, like hmLF, Helaina rhLF displayed well-folded globular structures in solution. Reconstructed solvent envelopes of Helaina rhLF, obtained through the SAXS analysis, demonstrated a remarkable fit with the reported crystalline structure of iron-bound native hmLF. Differential scanning calorimetry investigations into the thermal stability of Helaina rhLF revealed two distinct denaturation temperatures at 68.7 ± 0.9 °C and 91.9 ± 0.5 °C, consistently mirroring denaturation temperatures observed for apo- and holo-hmLF. Overall, Helaina rhLF differed from hmLF in the N-glycans they possessed; nevertheless, the characterization results affirmed that Helaina rhLF was of high purity and exhibited globular structures closely akin to that of hmLF.
Collapse
Affiliation(s)
- Xiaoning Lu
- Helaina Inc., 345 Park Avenue South, 5th Floor, New York, NY 10010, USA.
| | - Chad Cummings
- Helaina Inc., 345 Park Avenue South, 5th Floor, New York, NY 10010, USA.
| | - Udodili A Osuala
- Helaina Inc., 345 Park Avenue South, 5th Floor, New York, NY 10010, USA.
| | - Neela H Yennawar
- X-ray Crystallography Core Facility, The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Kevin E W Namitz
- X-ray Crystallography Core Facility, The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Brittney Hellner
- Helaina Inc., 345 Park Avenue South, 5th Floor, New York, NY 10010, USA.
| | | | - Ross D Peterson
- Helaina Inc., 345 Park Avenue South, 5th Floor, New York, NY 10010, USA.
| | - Anthony J Clark
- Helaina Inc., 345 Park Avenue South, 5th Floor, New York, NY 10010, USA.
| |
Collapse
|
3
|
Cong Y, Zhang Y, Han Y, Wu Y, Wang D, Zhang B. Recommendations for nutritional supplements for dry eye disease: current advances. Front Pharmacol 2024; 15:1388787. [PMID: 38873421 PMCID: PMC11169594 DOI: 10.3389/fphar.2024.1388787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 04/17/2024] [Indexed: 06/15/2024] Open
Abstract
Dry eye disease (DED) represents a prevalent ocular surface disease. The development of effective nutritional management strategies for DED is crucial due to its association with various factors such as inflammation, oxidative stress, deficiencies in polyunsaturated fatty acids (PUFAs), imbalanced PUFA ratios, and vitamin insufficiencies. Extensive research has explored the impact of oral nutritional supplements, varying in composition and dosage, on the symptoms of DED. The main components of these supplements include fish oils (Omega-3 fatty acids), vitamins, trace elements, and phytochemical extracts. Beyond these well-known nutrients, it is necessary to explore whether novel nutrients might contribute to more effective DED management. This review provides a comprehensive update on the therapeutic potential of nutrients and presents new perspectives for combination supplements in DED treatment.
Collapse
Affiliation(s)
| | | | | | | | | | - Bingjie Zhang
- Department of Ophthalmology, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
4
|
Li C, Lu Y, Wang J, Liu B, Szeto IMY, Zhang W, Bi R, Duan S, Quan R, Wang X, Li Y, Xiong W, Sun J, Sun Y. Immunoregulation of bovine lactoferrin together with osteopontin promotes immune system development and maturation. Food Funct 2024; 15:866-880. [PMID: 38165790 DOI: 10.1039/d3fo03515h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2024]
Abstract
The immune system of infants is partly weak and immature, and supplementation of infant formula can be of vital importance to boost the development of the immune system. Lactoferrin (LF) and osteopontin (OPN) are essential proteins in human milk with immunoregulation function. An increasing number of studies indicate that proteins have interactions with each other in milk, and our previous study found that a ratio of LF : OPN at 1 : 5 (w/w, denoted as LOP) had a synergistic effect on intestinal barrier protection. It remains unknown whether LOP can also exert a stronger effect on immunoregulation. Hence, we used an in vitro model of LPS-induced macrophage inflammation and in vivo models of LPS-induced intestinal inflammation and early life development. We showed that LOP increased the secretion of the granulocyte-macrophage colony-stimulating factor (132%), stem cell factor (167%) and interleukin-3 (176%) in bone marrow cells, as well as thymosin (155%) and interleukin-10 (161%) in the thymus, more than LF or OPN alone during development, and inhibited changes in immune cells and cytokines during the LPS challenge. In addition, analysis of the components of digested proteins in vitro revealed that differentially expressed peptides may provide immunoregulation. Lastly, LOP increased the abundance of Rikenellaceae, Muribaculum, Faecalibaculum, and Elisenbergiella in the cecum content. These results imply that LOP is a potential immunomodifier for infants and offers a new theoretical basis for infant formula innovation.
Collapse
Affiliation(s)
- Chuangang Li
- Key Laboratory of Functional Dairy, Co-Constructed by Ministry of Education and Beijing Municipality, College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China. @
| | - Yao Lu
- Key Laboratory of Functional Dairy, Co-Constructed by Ministry of Education and Beijing Municipality, College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China. @
| | - Jian Wang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China. @
| | - Biao Liu
- Key Laboratory of Functional Dairy, Co-Constructed by Ministry of Education and Beijing Municipality, College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China
- Inner Mongolia Yili Industrial Group Co. Ltd, Yili Maternal and Infant Nutrition Institute (YMINI), Beijing, China
| | - Ignatius Man-Yau Szeto
- Inner Mongolia Yili Industrial Group Co. Ltd, Yili Maternal and Infant Nutrition Institute (YMINI), Beijing, China
- Inner Mongolia Dairy Technology Research Institute Co. Ltd, Hohhot, China
- National Center of Technology Innovation for Dairy, Hohhot, China
| | - Wen Zhang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China. @
| | - Ran Bi
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China. @
| | - Sufang Duan
- Inner Mongolia Yili Industrial Group Co. Ltd, Yili Maternal and Infant Nutrition Institute (YMINI), Beijing, China
- Inner Mongolia Dairy Technology Research Institute Co. Ltd, Hohhot, China
| | - Rui Quan
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China. @
| | - Xuemin Wang
- Inner Mongolia Yili Industrial Group Co. Ltd, Yili Maternal and Infant Nutrition Institute (YMINI), Beijing, China
- Inner Mongolia Dairy Technology Research Institute Co. Ltd, Hohhot, China
| | - Yixuan Li
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China. @
| | - Wei Xiong
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China. @
- Food Laboratory of Zhongyuan, Luohe 462000, China
| | - Jiazeng Sun
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China. @
| | - Yanan Sun
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China. @
| |
Collapse
|
5
|
Li C, Sun Y, He T, Lu Y, Szeto IMY, Duan S, Zhang Y, Liu B, Zhang Y, Zhang W, He J, Li Y. Synergistic effect of lactoferrin and osteopontin on intestinal barrier injury. Int J Biol Macromol 2023; 253:127416. [PMID: 37838132 DOI: 10.1016/j.ijbiomac.2023.127416] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 10/10/2023] [Accepted: 10/10/2023] [Indexed: 10/16/2023]
Abstract
Several studies indicate that the disruption of the intestinal epithelial barrier can lead to inflammatory bowel disease (IBD). Recent evidence has increasingly demonstrated that lactoferrin (LF) and osteopontin (OPN) can alleviate intestinal barrier injury. However, the potential synergistic effects of these two proteins and the mechanisms underlying their effects remain unclear. To address this question, we developed a lipopolysaccharide-induced intestinal barrier injury model in C57BL/6 N mice. Our findings demonstrated that the combination of LF and OPN at a 1:5 ratio exerts the strongest protective effect on the intestinal barrier, and it is more effective than LF or OPN alone. This protection is evidenced by the decrease in serum diamine oxidase (DAO) activity (1.66-fold decrease) and D-lactic content (1.51-fold decrease) and the reduced rate of FITC-labeled glucan transport across the jejunum (3.18-fold decrease). Moreover, the protein combination significantly promoted villi length (1.66-fold increase) and crypt depth (1.57-fold increase), improved tight junction protein structure and expression, and boosted the number of absorptive cells (4.34-fold increase) in the intestinal epithelium. Furthermore, the combination promoted crypt cell proliferation and differentiation via Notch signaling. In summary, our findings provide scientific evidence supporting the use of dietary intervention strategies for preventing IBD.
Collapse
Affiliation(s)
- Chuangang Li
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China; Key Laboratory of Functional Daily, Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Yanan Sun
- Key Laboratory of Functional Daily, Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Tingchao He
- Inner Mongolia Dairy Technology Research Institute Co. Ltd., Hohhot 010110, China; Inner Mongolia Yili Industrial Group, Co. Ltd., Yili Maternal and Infant Nutrition Institute (YMINI), Beijing 100070, China
| | - Yao Lu
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Ignatius Man-Yau Szeto
- Inner Mongolia Yili Industrial Group, Co. Ltd., Yili Maternal and Infant Nutrition Institute (YMINI), Beijing 100070, China; National Center of Technology Innovation for Dairy, Hohhot 010110, China
| | - Sufang Duan
- Inner Mongolia Dairy Technology Research Institute Co. Ltd., Hohhot 010110, China; Inner Mongolia Yili Industrial Group, Co. Ltd., Yili Maternal and Infant Nutrition Institute (YMINI), Beijing 100070, China
| | - Yifan Zhang
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Biao Liu
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China; Inner Mongolia Yili Industrial Group, Co. Ltd., Yili Maternal and Infant Nutrition Institute (YMINI), Beijing 100070, China
| | - Yiran Zhang
- Key Laboratory of Functional Daily, Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Wen Zhang
- Key Laboratory of Functional Daily, Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Jian He
- National Center of Technology Innovation for Dairy, Hohhot 010110, China
| | - Yixuan Li
- Key Laboratory of Functional Daily, Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
6
|
Alamdari-Palangi V, Jaberi KR, Shahverdi M, Naeimzadeh Y, Tajbakhsh A, Khajeh S, Razban V, Fallahi J. Recent advances and applications of peptide-agent conjugates for targeting tumor cells. J Cancer Res Clin Oncol 2023; 149:15249-15273. [PMID: 37581648 DOI: 10.1007/s00432-023-05144-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 07/08/2023] [Indexed: 08/16/2023]
Abstract
BACKGROUND Cancer, being a complex disease, presents a major challenge for the scientific and medical communities. Peptide therapeutics have played a significant role in different medical practices, including cancer treatment. METHOD This review provides an overview of the current situation and potential development prospects of anticancer peptides (ACPs), with a particular focus on peptide vaccines and peptide-drug conjugates for cancer treatment. RESULTS ACPs can be used directly as cytotoxic agents (molecularly targeted peptides) or can act as carriers (guiding missile) of chemotherapeutic agents and radionuclides by specifically targeting cancer cells. More than 60 natural and synthetic cationic peptides are approved in the USA and other major markets for the treatment of cancer and other diseases. Compared to traditional cancer treatments, peptides exhibit anticancer activity with high specificity and the ability to rapidly kill target cancer cells. ACP's target and kill cancer cells via different mechanisms, including membrane disruption, pore formation, induction of apoptosis, necrosis, autophagy, and regulation of the immune system. Modified peptides have been developed as carriers for drugs, vaccines, and peptide-drug conjugates, which have been evaluated in various phases of clinical trials for the treatment of different types of solid and leukemia cancer. CONCLUSIONS This review highlights the potential of ACPs as a promising therapeutic option for cancer treatment, particularly through the use of peptide vaccines and peptide-drug conjugates. Despite the limitations of peptides, such as poor metabolic stability and low bioavailability, modified peptides show promise in addressing these challenges. Various mechanism of action of anticancer peptides. Modes of action against cancer cells including: inducing apoptosis by cytochrome c release, direct cell membrane lysis (necrosis), inhibiting angiogenesis, inducing autophagy-mediated cell death and immune cell regulation.
Collapse
Affiliation(s)
- Vahab Alamdari-Palangi
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, 7133654361, Iran
| | - Khojaste Rahimi Jaberi
- Department of Neuroscience, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahshid Shahverdi
- Medical Biotechnology Research Center, Arak University of Medical Sciences, Arak, Iran
| | - Yasaman Naeimzadeh
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, 7133654361, Iran
| | - Amir Tajbakhsh
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, 7133654361, Iran
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sahar Khajeh
- Bone and Joint Diseases Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Vahid Razban
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, 7133654361, Iran.
| | - Jafar Fallahi
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, 7133654361, Iran.
| |
Collapse
|
7
|
Conesa C, Bellés A, Grasa L, Sánchez L. The Role of Lactoferrin in Intestinal Health. Pharmaceutics 2023; 15:1569. [PMID: 37376017 DOI: 10.3390/pharmaceutics15061569] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 05/16/2023] [Accepted: 05/19/2023] [Indexed: 06/29/2023] Open
Abstract
The intestine represents one of the first barriers where microorganisms and environmental antigens come into tight contact with the host immune system. A healthy intestine is essential for the well-being of humans and animals. The period after birth is a very important phase of development, as the infant moves from a protected environment in the uterus to one with many of unknown antigens and pathogens. In that period, mother's milk plays an important role, as it contains an abundance of biologically active components. Among these components, the iron-binding glycoprotein, lactoferrin (LF), has demonstrated a variety of important benefits in infants and adults, including the promotion of intestinal health. This review article aims to provide a compilation of all the information related to LF and intestinal health, in infants and adults.
Collapse
Affiliation(s)
- Celia Conesa
- Departamento de Producción Animal y Ciencia de los Alimentos, Facultad de Veterinaria, Universidad de Zaragoza, 50013 Zaragoza, Spain
| | - Andrea Bellés
- Departamento de Farmacología, Fisiología y Medicina Legal y Forense, Facultad de Veterinaria, Universidad de Zaragoza, 50013 Zaragoza, Spain
- Instituto Agroalimentario de Aragón IA2 (UNIZAR-CITA), 50013 Zaragoza, Spain
| | - Laura Grasa
- Departamento de Farmacología, Fisiología y Medicina Legal y Forense, Facultad de Veterinaria, Universidad de Zaragoza, 50013 Zaragoza, Spain
- Instituto Agroalimentario de Aragón IA2 (UNIZAR-CITA), 50013 Zaragoza, Spain
- Instituto de Investigación Sanitaria de Aragón (IIS Aragón), 50009 Zaragoza, Spain
| | - Lourdes Sánchez
- Departamento de Producción Animal y Ciencia de los Alimentos, Facultad de Veterinaria, Universidad de Zaragoza, 50013 Zaragoza, Spain
- Instituto Agroalimentario de Aragón IA2 (UNIZAR-CITA), 50013 Zaragoza, Spain
| |
Collapse
|
8
|
Han B, Zhang L, Zhou P. Comparative proteomics of whey proteins: New insights into quantitative differences between bovine, goat and camel species. Int J Biol Macromol 2023; 227:10-16. [PMID: 36529209 DOI: 10.1016/j.ijbiomac.2022.12.103] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 12/10/2022] [Indexed: 12/23/2022]
Abstract
Whey proteins are the leading proteins class in milk and play an essential role in the immune defense of neonatal mammals. The aim of this study was to analyze whey proteins in bovine, goat and camel milk by label free proteomics techniques. Finally, 840 proteins were identified, which considerably increasing the number of whey proteins identified in these species. The results of the PCA revealed significant differences in whey proteome patterns between bovine, goat and camel milk. Proteins such as PAEP, CST3, SERPING1, CTSB and GLG1 play an important role as markers in the classification of bovine, goat and camel milk. Statistical analysis showed that the relative abundances of many whey proteins such as ALB, LALBA, LTF and LPO were significantly different among different species. GO and KEGG functional analysis have shown that while the distribution of biological functions involved in whey proteins was relatively similar across species, they differed in terms of protein quantity. These data shed light on the quantitative differences and potential physiological functions of whey proteins across species, and may point the way to the production of specific functional whey proteins.
Collapse
Affiliation(s)
- Binsong Han
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Lina Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Peng Zhou
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China.
| |
Collapse
|
9
|
Kharouba M, El-Kamel A, Mehanna R, Thabet E, Heikal L. Pitavastatin-loaded bilosomes for oral treatment of hepatocellular carcinoma: a repurposing approach. Drug Deliv 2022; 29:2925-2944. [PMID: 36081339 PMCID: PMC9467608 DOI: 10.1080/10717544.2022.2120925] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Albeit its established efficacy as an anti-hyperlipidemic agent, pitavastatin (PIT) has been shown to have other various therapeutic effects. One of these effects is the anti-cancer activity against hepatocellular carcinoma (HCC). This effect has been evaluated in this study for the first time via its oral delivery loaded in bilosomes both in vitro in hepatocellular carcinoma (HCC) cell line; HepG2 and in vivo in an Ehrlich ascites carcinoma (EAC) model. Moreover, the impact of surface modification of bilosomes with lactoferrin (LF) as an active targeting ligand for HCC was investigated. Bilosomes were prepared by thin-film hydration and different molar phospholipid to bile salt ratios were used to optimize the bilosomal formulation. The molar phospholipid to bile salt ratio was adjusted to 4:1 at pH 7.4. LF-coated bilosomes possessed a particle size, PDI, entrapment efficiency, and zeta potential of 112.28 nm ± 6.35, 0.229 ± 0.06, 90.56% ± 3.22, and −7.86 mV ± 1.13, respectively. LF-coated bilosomes also increased permeation of PIT when tested on Caco-2 cells by 3.1-folds (compared to uncoated ones or free PIT solution). It also improved the cytotoxicity of HepG2 spheroids 44-folds more than PIT-free solution. RT-PCR analysis showed that LF-coated PIT-loaded bilosomes caused an improvement (2-fold increase) in the apoptotic potential of PIT mediated by caspase-3. In conclusion, the optimized LF-coated PIT-loaded bilosomes were cytotoxic to HCC with improved hepatocytes permeation and cellular uptake. Thus, the proposed formula could be a promising treatment for HCC.
Collapse
Affiliation(s)
- Maged Kharouba
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Amal El-Kamel
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Radwa Mehanna
- Medical Physiology Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt.,Center of Excellence for Research in Regenerative Medicine and its Applications CERRMA, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Eman Thabet
- Medical Physiology Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt.,Center of Excellence for Research in Regenerative Medicine and its Applications CERRMA, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Lamia Heikal
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| |
Collapse
|
10
|
Jia SN, Han YB, Yang R, Yang ZC. Chemokines in colon cancer progression. Semin Cancer Biol 2022; 86:400-407. [PMID: 35183412 DOI: 10.1016/j.semcancer.2022.02.007] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/02/2022] [Accepted: 02/03/2022] [Indexed: 01/27/2023]
Abstract
Colon cancer is a major human cancer accounting for about a tenth of all cancer cases thus making it among the top three cancers in terms of incidence as well as mortality. Metastasis to distant organs, particularly to liver, is the primary reason for associated mortality. Chemokines, the chemo-attractants for various immune cells, have increasingly been reported to be involved in cancer initiation and progression, including in colon cancer. Here we discuss the available knowledge on the role of several chemokines, such as, CCL2, CCL3, CCL5, CXCL1, CXCL2, CXCL8 in colon cancer progression. CCL20 is one chemokine with emerging evidence for its role in influencing colon cancer tumor microenvironment through the documents effects on fibroblasts, macrophages and immune cells. We focus on CCL20 and its receptor CCR6 as promising factors that affect multiple levels of colon cancer progression. They interact with several cytokines and TLR receptors leading to increased aggressiveness, as supported by multitude of evidence from in vitro, in vivo studies as well as human patient samples. CCL20-CCR6 bring about their biological effects through regulation of several signaling pathways, including, ERK and NF-κB pathways, in addition to the epithelial-mesenchymal transition. Signaling involving CCL20-CCR6 has profound effect on colon cancer hepatic metastasis. Combined with elevated CCL20 levels in colon tumors and metastatic patients, the above information points to a need for further evaluation of chemokines as diagnostic and/or prognostic biomarkers.
Collapse
Affiliation(s)
- Sheng-Nan Jia
- Department of HepatoPancreatoBiliary Medicine, The Second Hospital of Jilin University, Changchun, 130000, China
| | - Ying-Bo Han
- Department of Gastrointestinal Surgery, The Second Hospital of Jilin University, Changchun, 130000, China
| | - Rui Yang
- Department of Gastroenterology, The Second Hospital of Jilin University, Changchun, 130000, China
| | - Ze-Cheng Yang
- Department of Gastrointestinal Surgery, The Second Hospital of Jilin University, Changchun, 130000, China.
| |
Collapse
|
11
|
Lath A, Santal AR, Kaur N, Kumari P, Singh NP. Anti-cancer peptides: their current trends in the development of peptide-based therapy and anti-tumor drugs. Biotechnol Genet Eng Rev 2022; 39:45-84. [PMID: 35699384 DOI: 10.1080/02648725.2022.2082157] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Human cancer remains a cause of high mortality throughout the world. The conventional methods and therapies currently employed for treatment are followed by moderate-to-severe side effects. They have not generated curative results due to the ineffectiveness of treatments. Besides, the associated high costs, technical requirements, and cytotoxicity further characterize their limitations. Due to relatively higher presidencies, bioactive peptides with anti-cancer attributes have recently become treatment choices within the therapeutic arsenal. The peptides act as potential anti-cancer agents explicitly targeting tumor cells while being less toxic to normal cells. The anti-cancer peptides are isolated from various natural sources, exhibit high selectivity and high penetration efficiency, and could be quickly restructured. The therapeutic benefits of compatible anti-cancer peptides have contributed to the significant expansion of cancer treatment; albeit, the mechanisms by which bioactive peptides inhibit the proliferation of tumor cells remain unclear. This review will provide a framework for assessing anti-cancer peptides' structural and functional aspects. It shall provide appropriate information on their mode of action to support and strengthen efforts to improve cancer prevention. The article will mention the therapeutic health benefits of anti-cancer peptides. Their importance in clinical studies is elaborated for reducing cancer incidences and developing sustainable treatment models.
Collapse
Affiliation(s)
- Amit Lath
- Centre for Biotechnology, Maharshi Dayanand University, Rohtak, India
| | - Anita Rani Santal
- Department of Microbiology, Maharshi Dayanand University, Rohtak, India
| | - Nameet Kaur
- Amity Institute of Biotechnology, Amity University, Noida, India
| | - Poonam Kumari
- Sophisticated Analytical Instrumentation Facility, CIL and UCIM, Punjab University, Chandigarh, Inida
| | - Nater Pal Singh
- Centre for Biotechnology, Maharshi Dayanand University, Rohtak, India
| |
Collapse
|
12
|
Wells JM, Gao Y, de Groot N, Vonk MM, Ulfman L, van Neerven RJJ. Babies, Bugs, and Barriers: Dietary Modulation of Intestinal Barrier Function in Early Life. Annu Rev Nutr 2022; 42:165-200. [PMID: 35697048 DOI: 10.1146/annurev-nutr-122221-103916] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The intestinal barrier is essential in early life to prevent infection, inflammation, and food allergies. It consists of microbiota, a mucus layer, an epithelial layer, and the immune system. Microbial metabolites, the mucus, antimicrobial peptides, and secretory immunoglobulin A (sIgA) protect the intestinal mucosa against infection. The complex interplay between these functionalities of the intestinal barrier is crucial in early life by supporting homeostasis, development of the intestinal immune system, and long-term gut health. Exclusive breastfeeding is highly recommended during the first 6 months. When breastfeeding is not possible, milk-based infant formulas are the only safe alternative. Breast milk contains many bioactive components that help to establish the intestinal microbiota and influence the development of the intestinal epithelium and the immune system. Importantly, breastfeeding lowers the risk for intestinal and respiratory tract infections. Here we review all aspects of intestinal barrier function and the nutritional components that impact its functionality in early life, such as micronutrients, bioactive milk proteins, milk lipids, and human milk oligosaccharides. These components are present in breast milk and can be added to milk-based infant formulas to support gut health and immunity. Expected final online publication date for the Annual Review of Nutrition, Volume 42 is August 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Jerry M Wells
- Host Microbe Interactomics, Wageningen University and Research, Wageningen, The Netherlands
| | - Yifan Gao
- Cell Biology and Immunology, Wageningen University and Research, Wageningen, The Netherlands
| | | | | | | | - R J Joost van Neerven
- Cell Biology and Immunology, Wageningen University and Research, Wageningen, The Netherlands.,FrieslandCampina, Amersfoort, The Netherlands;
| |
Collapse
|
13
|
Pan M, Yang J, Liu K, Xie X, Hong L, Wang S, Wang S. Irradiation technology: An effective and promising strategy for eliminating food allergens. Food Res Int 2021; 148:110578. [PMID: 34507726 DOI: 10.1016/j.foodres.2021.110578] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 06/23/2021] [Accepted: 06/27/2021] [Indexed: 11/29/2022]
Abstract
Food allergies are one of the major health concerns worldwide and have been increasing at an alarming rate in recent times. The elimination of food allergenicity has been an important issue in current research on food. Irradiation is a typical nonthermal treatment technology that can effectively reduce the allergenicity of food, showing great application prospects in improving the quality and safety of foods. In this review, the mechanism and remarkable features of irradiation in the elimination of food allergens are mainly introduced, and the research progress on reducing the allergenicity of animal foods (milk, egg, fish and shrimp) and plant foods (soybean, peanut, wheat and nuts) using irradiation is summarized. Furthermore, the influencing factors for irradiation in the elimination of food allergens are analyzed and further research directions of irradiation desensitization technology are also discussed. This article aims to provide a reference for promoting the application of irradiation technology in improving the safety of foods.
Collapse
Affiliation(s)
- Mingfei Pan
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science and Technology, Tianjin 300457, China; Key Laboratory of Food Nutrition and Safety, Ministry of Education of China, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Jingying Yang
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science and Technology, Tianjin 300457, China; Key Laboratory of Food Nutrition and Safety, Ministry of Education of China, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Kaixin Liu
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science and Technology, Tianjin 300457, China; Key Laboratory of Food Nutrition and Safety, Ministry of Education of China, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Xiaoqian Xie
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science and Technology, Tianjin 300457, China; Key Laboratory of Food Nutrition and Safety, Ministry of Education of China, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Liping Hong
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science and Technology, Tianjin 300457, China; Key Laboratory of Food Nutrition and Safety, Ministry of Education of China, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Shan Wang
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science and Technology, Tianjin 300457, China; Key Laboratory of Food Nutrition and Safety, Ministry of Education of China, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Shuo Wang
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science and Technology, Tianjin 300457, China; Key Laboratory of Food Nutrition and Safety, Ministry of Education of China, Tianjin University of Science and Technology, Tianjin 300457, China.
| |
Collapse
|
14
|
Nayak A, Tiozzo C, Lin X, Mejia C, Gurzenda E, Kim M, Hanna N. Is Lactoferrin Supplementation Beneficial for All Preterm Infants? Am J Perinatol 2021; 40:680-687. [PMID: 34058763 DOI: 10.1055/s-0041-1730433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
OBJECTIVE Human milk (HM) has antibacterial properties due to the presence of immune-modulators, including lactoferrin (LF). This study will determine effect(s) of HM maturation, fortification, and storage conditions on LF levels and its antibacterial properties. STUDY DESIGN HM samples (n = 30) were obtained from preterm and term mothers. The LF levels were analyzed by ELISA, and the antibacterial activity was measured after inoculation with Escherichia coli. RESULTS The highest level of LF in preterm HM was observed in the first week of lactation. However, storage of preterm HM at 4°C decreased LF levels significantly. Both LF levels and antibacterial activity in preterm HM was lower compared with term HM, but significantly higher than donor HM even after HM-based fortification. LF supplementation of donor HM improved its antibacterial activity. CONCLUSION Preterm infants fed donor HM, formula, or stored HM at 4°C may benefits from LF supplementation to improve HM antibacterial properties. KEY POINTS · Milk LF levels vary with storage and maturity.. · Donor milk is deficient in LF even after adding HM-based fortification.. · Donor HM and formula fed infants may benefit from LF..
Collapse
Affiliation(s)
- Amrita Nayak
- Department of Pediatrics, NYU Langone Hospital-Long Island, Mineola, New York
| | - Caterina Tiozzo
- Department of Pediatrics, NYU Langone Hospital-Long Island, Mineola, New York.,Women and Children Research Laboratory, NYU Long Island School of Medicine, Mineola, New York
| | - Xinhua Lin
- Women and Children Research Laboratory, NYU Long Island School of Medicine, Mineola, New York
| | - Claudia Mejia
- Women and Children Research Laboratory, NYU Long Island School of Medicine, Mineola, New York
| | - Ellen Gurzenda
- Women and Children Research Laboratory, NYU Long Island School of Medicine, Mineola, New York
| | - Maureen Kim
- Department of Pediatrics, NYU Langone Hospital-Long Island, Mineola, New York
| | - Nazeeh Hanna
- Department of Pediatrics, NYU Langone Hospital-Long Island, Mineola, New York.,Women and Children Research Laboratory, NYU Long Island School of Medicine, Mineola, New York
| |
Collapse
|
15
|
Biernbaum EN, Gnezda A, Akbar S, Franklin R, Venturelli PA, McKillip JL. Lactoferrin as an antimicrobial against Salmonella enterica and Escherichia coli O157:H7 in raw milk. JDS COMMUNICATIONS 2021; 2:92-97. [PMID: 36339505 PMCID: PMC9623761 DOI: 10.3168/jdsc.2020-0030] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 01/30/2021] [Indexed: 06/16/2023]
Abstract
Improper storage conditions or processing of milk leads to potential spoilage and illness, due in part to temperature abuse, allowing bacteria present to grow and spoil the product. However, certain proteins naturally found in raw milk, such as lactoferrin, have reported antibacterial properties. The levels of lactoferrin required to effectively inhibit growth of pathogens have not been investigated thoroughly. This study aimed to examine various concentrations of lactoferrin as a potential biopreservative and as an antimicrobial against the common dairy pathogens Salmonella enterica and Escherichia coli O157:H7. Minimum inhibitory concentration assays were conducted on raw bovine milk in which the bacteria were exposed to varying concentrations of lactoferrin. In the raw milk system, the growth of E. coli O157:H7 was significantly decreased at levels greater than 14.05 mg/mL lactoferrin based on the reduction of tetrazolium salts. For S. enterica, only lactoferrin concentrations at or above 112.5 mg/mL in the milk resulted in reduced growth. Taken together, these results indicate that lactoferrin may have biopreservative potential. To fully examine the practicality and effectiveness of lactoferrin as an antimicrobial additive, a similar study should be conducted using additional (gram-positive) pathogens, such as Bacillus cereus and Listeria monocytogenes. If effective, lactoferrin could prolong the shelf life of dairy products and help reduce the incidence of foodborne illnesses in developing countries with limited refrigeration capability.
Collapse
Affiliation(s)
| | - Anita Gnezda
- Department of Chemistry, Ball State University, Muncie, IN 47306
| | - Samina Akbar
- Department of Biomedical Sciences, Marian University, Indianapolis, IN 46222
| | - Rose Franklin
- Department of Biology, Ball State University, Muncie, IN 47306
| | | | | |
Collapse
|
16
|
Mikulic N, Uyoga MA, Mwasi E, Stoffel NU, Zeder C, Karanja S, Zimmermann MB. Iron Absorption is Greater from Apo-Lactoferrin and is Similar Between Holo-Lactoferrin and Ferrous Sulfate: Stable Iron Isotope Studies in Kenyan Infants. J Nutr 2020; 150:3200-3207. [PMID: 32886113 DOI: 10.1093/jn/nxaa226] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 04/14/2020] [Accepted: 07/09/2020] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Whether lactoferrin (Lf) binds iron to facilitate its absorption or to sequester iron from potential enteropathogens remains uncertain. Bovine Lf is added to many infant formulas, but previous studies in infants reported that Lf had no effect on or inhibited iron absorption. The effects of the apo (iron-free) or the holo (iron-loaded) forms of Lf on iron absorption are unclear. OBJECTIVES Our objective was to compare iron absorption from a maize-based porridge containing: 1) labeled ferrous sulfate (FeSO4) alone; 2) labeled FeSO4 given with bovine apo-Lf; and 3) intrinsically labeled bovine holo-Lf. METHODS In a crossover study, we measured iron absorption in Kenyan infants (n = 25; mean ± SD age 4.2 ± 0.9 months; mean ± SD hemoglobin 109 ± 11 g/L) from maize-based test meals containing: 1) 1.5 mg of iron as 54Fe-labeled FeSO4; 2) 1.42 mg of iron as 58Fe-labeled FeSO4, given with 1.41 g apo-Lf (containing 0.08 mg iron); and 3) 1.41 g holo-Lf carrying 1.5 mg iron as 57Fe. The iron saturation levels of apo- and holo-Lf were 0.56% and 47.26%, respectively primary outcome was fractional iron absorption (FIA), assessed by erythrocyte incorporation of isotopic labels. RESULTS The FIA from the meal containing apo-Lf + FeSO4 (geometric mean, 9.8%; -SD and +SD, 5.4% and 17.5%) was higher than from the meals containing FeSO4 (geometric mean, 6.3%; -SD and +SD, 3.2% and 12.6%; P = 0.002) or holo-Lf (geometric mean, 5.0%; -SD and +SD, 2.8% and 8.9%; P <0.0001). There was no significant difference in FIA when comparing the meals containing holo-Lf versus FeSO4 alone (P = 0.24). CONCLUSIONS The amount of iron absorbed from holo-Lf was comparable to that of FeSO4, and the addition of apo-Lf to a test meal containing FeSO4 significantly increased (+56%) iron absorption. These findings suggest that Lf facilitates iron absorption in young infants. Because Lf binds iron with high affinity, it could be a safe way to provide iron to infants in low-income countries, where iron fortificants can adversely affect the gut microbiome and cause diarrhea. This study was registered at clinicaltrials.gov as NCT03617575.
Collapse
Affiliation(s)
- Nadja Mikulic
- Laboratory of Human Nutrition, Institute of Food, Nutrition and Health, Swiss Federal Institute of Technology (ETH), Zurich, Switzerland
| | - Mary A Uyoga
- Laboratory of Human Nutrition, Institute of Food, Nutrition and Health, Swiss Federal Institute of Technology (ETH), Zurich, Switzerland
| | - Edith Mwasi
- Pediatrics Department, Msambweni County Referral Hospital, Msambweni, Kenya
| | - Nicole U Stoffel
- Laboratory of Human Nutrition, Institute of Food, Nutrition and Health, Swiss Federal Institute of Technology (ETH), Zurich, Switzerland
| | - Christophe Zeder
- Laboratory of Human Nutrition, Institute of Food, Nutrition and Health, Swiss Federal Institute of Technology (ETH), Zurich, Switzerland
| | - Simon Karanja
- Public and Community Health Department, Jomo Kenyatta University of Agriculture and Technology, Nairobi, Kenya
| | - Michael B Zimmermann
- Laboratory of Human Nutrition, Institute of Food, Nutrition and Health, Swiss Federal Institute of Technology (ETH), Zurich, Switzerland
| |
Collapse
|
17
|
Elzoghby AO, Abdelmoneem MA, Hassanin IA, Abd Elwakil MM, Elnaggar MA, Mokhtar S, Fang JY, Elkhodairy KA. Lactoferrin, a multi-functional glycoprotein: Active therapeutic, drug nanocarrier & targeting ligand. Biomaterials 2020; 263:120355. [PMID: 32932142 PMCID: PMC7480805 DOI: 10.1016/j.biomaterials.2020.120355] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 08/18/2020] [Accepted: 08/31/2020] [Indexed: 12/21/2022]
Abstract
Recent progress in protein-based nanomedicine, inspired by the success of Abraxane® albumin-paclitaxel nanoparticles, have resulted in novel therapeutics used for treatment of challenging diseases like cancer and viral infections. However, absence of specific drug targeting, poor pharmacokinetics, premature drug release, and off-target toxicity are still formidable challenges in the clinic. Therefore, alternative protein-based nanomedicines were developed to overcome those challenges. In this regard, lactoferrin (Lf), a glycoprotein of transferrin family, offers a promising biodegradable well tolerated material that could be exploited both as an active therapeutic and drug nanocarrier. This review highlights the major pharmacological actions of Lf including anti-cancer, antiviral, and immunomodulatory actions. Delivery technologies of Lf to improve its pries and enhance its efficacy were also reviewed. Moreover, different nano-engineering strategies used for fabrication of drug-loaded Lf nanocarriers were discussed. In addition, the use of Lf for functionalization of drug nanocarriers with emphasis on tumor-targeted drug delivery was illustrated. Besides its wide application in oncology nano-therapeutics, we discussed the recent advances of Lf-based nanocarriers as efficient platforms for delivery of anti-parkinsonian, anti-Alzheimer, anti-viral drugs, immunomodulatory and bone engineering applications.
Collapse
Affiliation(s)
- Ahmed O Elzoghby
- Center for Engineered Therapeutics, Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA; Harvard-MIT Division of Health Sciences & Technology (HST), Cambridge, MA, 02139, USA; Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt; Department of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt.
| | - Mona A Abdelmoneem
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt; Department of Pharmaceutics, Faculty of Pharmacy, Damanhur University, Damanhur, 22516, Egypt
| | - Islam A Hassanin
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt; Department of Biotechnology, Institute of Graduate Studies and Research, Alexandria University, Alexandria, 21526, Egypt
| | - Mahmoud M Abd Elwakil
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt; Laboratory of Innovative Nanomedicine, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo, 060-0812, Japan
| | - Manar A Elnaggar
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt; Nanotechnology Program, School of Sciences & Engineering, The American University in Cairo (AUC), New Cairo, 11835, Egypt
| | - Sarah Mokhtar
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt; Department of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt
| | - Jia-You Fang
- Pharmaceutics Laboratory, Graduate Institute of Natural Products, Chang Gung University, Taoyuan, 333, Taiwan; Research Center for Industry of Human Ecology, Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Kweishan, Taoyuan, 333, Taiwan; Department of Anesthesiology, Chang Gung Memorial Hospital, Kweishan, Taoyuan, 333, Taiwan
| | - Kadria A Elkhodairy
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt; Department of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt
| |
Collapse
|
18
|
Chiangjong W, Chutipongtanate S, Hongeng S. Anticancer peptide: Physicochemical property, functional aspect and trend in clinical application (Review). Int J Oncol 2020; 57:678-696. [PMID: 32705178 PMCID: PMC7384845 DOI: 10.3892/ijo.2020.5099] [Citation(s) in RCA: 168] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 06/26/2020] [Indexed: 01/10/2023] Open
Abstract
Cancer is currently ineffectively treated using therapeutic drugs, and is also able to resist drug action, resulting in increased side effects following drug treatment. A novel therapeutic strategy against cancer cells is the use of anticancer peptides (ACPs). The physicochemical properties, amino acid composition and the addition of chemical groups on the ACP sequence influences their conformation, net charge and orientation of the secondary structure, leading to an effect on targeting specificity and ACP-cell interaction, as well as peptide penetrating capability, stability and efficacy. ACPs have been developed from both naturally occurring and modified peptides by substituting neutral or anionic amino acid residues with cationic amino acid residues, or by adding a chemical group. The modified peptides lead to an increase in the effectiveness of cancer therapy. Due to this effectiveness, ACPs have recently been improved to form drugs and vaccines, which have sequentially been evaluated in various phases of clinical trials. The development of the ACPs remains focused on generating newly modified ACPs for clinical application in order to decrease the incidence of new cancer cases and decrease the mortality rate. The present review could further facilitate the design of ACPs and increase efficacious ACP therapy in the near future.
Collapse
Affiliation(s)
- Wararat Chiangjong
- Pediatric Translational Research Unit, Department of Pediatrics, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| | - Somchai Chutipongtanate
- Pediatric Translational Research Unit, Department of Pediatrics, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| | - Suradej Hongeng
- Division of Hematology and Oncology, Department of Pediatrics, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| |
Collapse
|
19
|
Prevention of Nosocomial Infections in Critically Ill Patients With Lactoferrin: A Randomized, Double-Blind, Placebo-Controlled Study. Crit Care Med 2019; 46:1450-1456. [PMID: 30015668 DOI: 10.1097/ccm.0000000000003294] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE To obtain preliminary evidence for the efficacy of lactoferrin as a preventative measure for nosocomial infections and inform the conduct of a definitive study. DESIGN Phase 2, multicenter, randomized, double-blind, placebo-controlled study. SETTING Medical-surgical ICUs. PATIENTS Adult, critically ill patients receiving invasive mechanical ventilation. INTERVENTIONS Randomized, eligible, consenting patients expected to require invasive mechanical ventilation more than 48 hours received lactoferrin both enterally and via an oral swab or a placebo of sterile water for up to 28 days. MEASUREMENTS AND MAIN RESULTS Of the 214 patients who were randomized, 212 received at least one dose of the intervention and were analyzed (107 lactoferrin and 105 placebo). Protocol adherence was 87.5%. Patients receiving lactoferrin were older (mean [SD], 66.3 [13.5] vs 62.5 [16.2] yr), had a higher Acute Physiology and Chronic Health Evaluation II score (26.8 [7.8] vs 23.5 [7.9]), and need for vasopressors (79% vs 70%). Antibiotic-free days (17.3 [9.0] vs 18.5 [7.1]; p = 0.91) and nosocomial infections (0.3 [0.7] vs 0.4 [0.6] per patient; p = 0.48) did not differ between lactoferrin and placebo groups, respectively. Clinical outcomes for lactoferrin versus placebo were as follows: ICU length of stay (14.5 [18.0] vs 15.0 [37.3] d; p = 0.82), hospital length of stay (25.0 [25.9] vs 28.1 [44.6] d; p = 0.57), hospital mortality (41.1% vs 30.5%; p = 0.11), and 90-day mortality (44.9% vs 32.4%; p = 0.06). Biomarker levels did not differ between the groups. CONCLUSIONS Lactoferrin did not improve the primary outcome of antibiotic-free days, nor any of the secondary outcomes. Our data do not support the conduct of a larger phase 3 trial.
Collapse
|
20
|
Assessment of bioactivities of the human milk lactoferrin–osteopontin complex in vitro. J Nutr Biochem 2019; 69:10-18. [DOI: 10.1016/j.jnutbio.2019.03.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 01/28/2019] [Accepted: 03/14/2019] [Indexed: 02/02/2023]
|
21
|
Lu J, Zhang S, Liu L, Pang X, Ma C, Jiang S, Lv J. Comparative proteomics analysis of human and ruminant milk serum reveals variation in protection and nutrition. Food Chem 2018; 261:274-282. [DOI: 10.1016/j.foodchem.2018.04.065] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 04/13/2018] [Accepted: 04/18/2018] [Indexed: 01/11/2023]
|
22
|
Manzoni P, García Sánchez R, Meyer M, Stolfi I, Pugni L, Messner H, Cattani S, Betta PM, Memo L, Decembrino L, Bollani L, Rinaldi M, Fioretti M, Quercia M, Maule M, Tavella E, Mussa A, Tzialla C, Laforgia N, Mosca F, Magaldi R, Mostert M, Farina D. Exposure to Gastric Acid Inhibitors Increases the Risk of Infection in Preterm Very Low Birth Weight Infants but Concomitant Administration of Lactoferrin Counteracts This Effect. J Pediatr 2018; 193:62-67.e1. [PMID: 29198543 DOI: 10.1016/j.jpeds.2017.09.080] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 09/26/2017] [Accepted: 09/28/2017] [Indexed: 01/08/2023]
Abstract
OBJECTIVE To investigate whether exposure to inhibitors of gastric acidity, such as H2 blockers or proton pump inhibitors, can independently increase the risk of infections in very low birth weight (VLBW) preterm infants in the neonatal intensive care unit. STUDY DESIGN This is a secondary analysis of prospectively collected data from a multicenter, randomized controlled trial of bovine lactoferrin (BLF) supplementation (with or without the probiotic Lactobacillus rhamnosus GG) vs placebo in prevention of late-onset sepsis (LOS) and necrotizing enterocolitis (NEC) in preterm infants. Inhibitors of gastric acidity were used at the recommended dosages/schedules based on the clinical judgment of attending physicians. The distribution of days of inhibitors of gastric acidity exposure between infants with and without LOS/NEC was assessed. The mutually adjusted effects of birth weight, gestational age, duration of inhibitors of gastric acidity treatment, and exposure to BLF were controlled through multivariable logistic regression. Interaction between inhibitors of gastric acidity and BLF was tested; the effects of any day of inhibitors of gastric acidity exposure were then computed for BLF-treated vs -untreated infants. RESULTS Two hundred thirty-five of 743 infants underwent treatment with inhibitors of gastric acidity, and 86 LOS episodes occurred. After multivariate analysis, exposure to inhibitors of gastric acidity remained significantly and independently associated with LOS (OR, 1.03; 95% CI, 1.008-1.067; P = .01); each day of inhibitors of gastric acidity exposure conferred an additional 3.7% odds of developing LOS. Risk was significant for Gram-negative (P < .001) and fungal (P = .001) pathogens, but not for Gram-positive pathogens (P = .97). On the test for interaction, 1 additional day of exposure to inhibitors of gastric acidity conferred an additional 7.7% risk for LOS (P = .003) in BLF-untreated infants, compared with 1.2% (P = .58) in BLF-treated infants. CONCLUSION Exposure to inhibitors of gastric acidity is significantly associated with the occurrence of LOS in preterm VLBW infants. Concomitant administration of BLF counteracts this selective disadvantage. TRIAL REGISTRATION isrctn.org: ISRCTN53107700.
Collapse
Affiliation(s)
- Paolo Manzoni
- Neonatology and NICU, S Anna Hospital, AOU Città della Salute e della Scienza, Torino, Italy.
| | - Ruben García Sánchez
- Neonatology and NICU, Complejo Asistencial Universitario de Salamanca, Salamanca, Spain
| | - Michael Meyer
- Neonatology and NICU, Middlemore Hospital, Auckland, New Zealand
| | - Ilaria Stolfi
- Neonatology, Azienda Ospedaliera Universitaria Policlinico Umberto I, Rome, Italy
| | - Lorenza Pugni
- NICU, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Università degli Studi di Milano, Milan, Italy
| | - Hubert Messner
- Neonatology and NICU, Ospedale Regionale, Bolzano/Bozen, Italy
| | - Silvia Cattani
- NICU, University of Modena and Reggio Emilia, Modena, Italy
| | | | - Luigi Memo
- UOC di Pediatria e Patologia Neonatale, Ospedale San Martino, Belluno, Italy
| | - Lidia Decembrino
- Patologia Neonatale, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Lina Bollani
- Patologia Neonatale, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | | | - Maria Fioretti
- Neonatology and NICU, S Anna Hospital, AOU Città della Salute e della Scienza, Torino, Italy
| | - Michele Quercia
- Cancer Epidemiology Unit, University of Turin, Department of Medical Sciences, Torino, Italy
| | - Milena Maule
- Cancer Epidemiology Unit, University of Turin, Torino, Italy
| | - Elena Tavella
- Neonatology and NICU, S Anna Hospital, AOU Città della Salute e della Scienza, Torino, Italy
| | - Alessandro Mussa
- Neonatology and NICU, S Anna Hospital, AOU Città della Salute e della Scienza, Torino, Italy
| | - Chryssoula Tzialla
- Patologia Neonatale, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Nicola Laforgia
- Cancer Epidemiology Unit, University of Turin, Department of Medical Sciences, Torino, Italy
| | - Fabio Mosca
- NICU, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Università degli Studi di Milano, Milan, Italy
| | | | - Michael Mostert
- Department of Pediatrics, University of Turin, Torino, Italy
| | - Daniele Farina
- Neonatology and NICU, S Anna Hospital, AOU Città della Salute e della Scienza, Torino, Italy
| | | |
Collapse
|
23
|
Benefits of Lactoferrin, Osteopontin and Milk Fat Globule Membranes for Infants. Nutrients 2017; 9:nu9080817. [PMID: 28788066 PMCID: PMC5579611 DOI: 10.3390/nu9080817] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 07/23/2017] [Accepted: 07/24/2017] [Indexed: 12/11/2022] Open
Abstract
The provision of essential and non-essential amino acids for breast-fed infants is the major function of milk proteins. In addition, breast-fed infants might benefit from bioactivities of milk proteins, which are exhibited in the intestine during the digestive phase and by absorption of intact proteins or derived peptides. For lactoferrin, osteopontin and milk fat globule membrane proteins/lipids, which have not until recently been included in substantial amounts in infant formulas, in vitro experiments and animal models provide a convincing base of evidence for bioactivities, which contribute to the protection of the infant from pathogens, improve nutrient absorption, support the development of the immune system and provide components for optimal neurodevelopment. Technologies have become available to obtain these compounds from cow´s milk and the bovine compounds also exhibit bioactivities in humans. Randomized clinical trials with experimental infant formulas incorporating lactoferrin, osteopontin, or milk fat globule membranes have already provided some evidence for clinical benefits. This review aims to compare findings from laboratory and animal experiments with outcomes of clinical studies. There is good justification from basic science and there are promising results from clinical studies for beneficial effects of lactoferrin, osteopontin and the milk fat globule membrane complex of proteins and lipids. Further studies should ideally be adequately powered to investigate effects on clinically relevant endpoints in healthy term infants.
Collapse
|
24
|
Xia X, Liu H, Lv H, Zhang J, Zhou J, Zhao Z. Preparation, characterization, and in vitro/vivo studies of oleanolic acid-loaded lactoferrin nanoparticles. DRUG DESIGN DEVELOPMENT AND THERAPY 2017; 11:1417-1427. [PMID: 28533680 PMCID: PMC5431734 DOI: 10.2147/dddt.s133997] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Oleanolic acid (OA), a pentacyclic triterpene, is used to safely and economically treat hepatopathy. However, OA, a Biopharmaceutics Classification System IV category drug, has low bioavailability owing to low solubility (<1 μg/mL) and biomembrane permeability. We developed a novel OA nanoparticle (OA-NP)-loaded lactoferrin (Lf) nanodelivery system with enhanced in vitro OA dissolution and improved oral absorption and bioavailability. The OA-NPs were prepared using NP albumin-bound technology and characterized using dynamic light scattering, scanning electron microscopy, X-ray powder diffraction, differential scanning calorimetry, and in vitro dissolution test. The in vivo pharmacokinetics was investigated in Sprague Dawley rats using liquid chromatography-tandem mass spectrometry. OA-NPs (OA:Lf =1:6, w/w%) exhibited spherical morphology, 202.2±8.3 nm particle size, +(27.1±0.32) mV ζ potential, 92.59%±3.24% encapsulation efficiency, and desirable in vitro release profiles. An effective in vivo bioavailability (340.59%) was achieved compared to the free drug following oral administration to rats. The Lf novel nanodelivery vehicle enhanced the dissolution rate, intestinal absorption, and bioavailability of OA. These results demonstrate that Lf NPs are a new strategy for improving oral absorption and bioavailability of poorly soluble and poorly absorbed drugs.
Collapse
Affiliation(s)
- Xiaojing Xia
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing.,Department of Pharmaceutics, ZheJiang Pharmaceutical College, Ningbo
| | - Haowei Liu
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing
| | - Huixia Lv
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing
| | - Jing Zhang
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing
| | - Jianping Zhou
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing
| | - Zhiying Zhao
- Department of Traditional Chinese Medicine, China Pharmaceutical University, Nanjing, People's Republic of China
| |
Collapse
|
25
|
Aly E, López-Nicolás R, Darwish AA, Frontela-Saseta C, Ros-Berruezo G. Supplementation of infant formulas with recombinant human lactoferrin and/or galactooligosaccharides increases iron bioaccessibility as measured by ferritin formed in Caco-2 cell model. Food Res Int 2016. [DOI: 10.1016/j.foodres.2016.08.030] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
26
|
Sherman MP, Adamkin DH, Niklas V, Radmacher P, Sherman J, Wertheimer F, Petrak K. Randomized Controlled Trial of Talactoferrin Oral Solution in Preterm Infants. J Pediatr 2016; 175:68-73.e3. [PMID: 27260839 PMCID: PMC4981514 DOI: 10.1016/j.jpeds.2016.04.084] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 03/22/2016] [Accepted: 04/25/2016] [Indexed: 11/30/2022]
Abstract
OBJECTIVE To evaluate the safety and explore the efficacy of recombinant human lactoferrin (talactoferrin [TLf]) to reduce infection. STUDY DESIGN We conducted a randomized, double blind, placebo-controlled trial in infants with birth weight of 750-1500 g. Infants received enteral TLf (n = 60) or placebo (n = 60) on days 1 through 28 of life; the TLf dose was 150 mg/kg every 12 hours. Primary outcomes were bacteremia, pneumonia, urinary tract infection, meningitis, and necrotizing enterocolitis (NEC). Secondary outcomes were sepsis syndrome and suspected NEC. We recorded clinical, laboratory, and radiologic findings, along with diseases and adverse events, in a database used for statistical analyses. RESULTS Demographic data were similar in the 2 groups of infants. We attributed no enteral or organ-specific adverse events to TLf. There were 2 deaths in the TLf group (1 each due to posterior fossa hemorrhage and postdischarge sudden infant death), and 1 death in the placebo group, due to NEC. The rate of hospital-acquired infections was 50% lower in the TLf group compared with the placebo group (P < .04), including fewer blood or line infections, urinary tract infections, and pneumonia. Fourteen infants in the TLf group weighing <1 kg at birth had no gram-negative infections, compared with only 3 of 14 such infants in the placebo group. Noninfectious outcomes were not statistically significantly different between the 2 groups, and there were no between-group differences in growth or neurodevelopment over a 1-year posthospitalization period. CONCLUSION We found no clinical or laboratory toxicity and a trend toward less infectious morbidity in the infants treated with TLf. TRIAL REGISTRATION ClinicalTrials.gov: NCT00854633.
Collapse
Affiliation(s)
- Michael P Sherman
- Division of Neonatology, Department of Child Health, University of Missouri, Columbia, MO.
| | - David H Adamkin
- Division of Neonatal Medicine, Department of Pediatrics, University of Louisville
| | - Victoria Niklas
- Division of Neonatal Medicine, Children’s Hospital Los Angeles, Keck School of Medicine at the University of Southern California, Los Angeles, California
| | - Paula Radmacher
- Division of Neonatal Medicine, Department of Pediatrics, University of Louisville
| | - Jan Sherman
- Division of Neonatology, Department of Child Health, University of Missouri - Columbia,Sinclair School of Nursing, University of Missouri, Columbia, Missouri
| | - Fiona Wertheimer
- Division of Neonatal Medicine, Children’s Hospital Los Angeles, Keck School of Medicine at the University of Southern California, Los Angeles, California
| | | |
Collapse
|
27
|
Lönnerdal B, Erdmann P, Thakkar SK, Sauser J, Destaillats F. Longitudinal evolution of true protein, amino acids and bioactive proteins in breast milk: a developmental perspective. J Nutr Biochem 2016; 41:1-11. [PMID: 27771491 DOI: 10.1016/j.jnutbio.2016.06.001] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 06/07/2016] [Indexed: 12/11/2022]
Abstract
The protein content of breast milk provides a foundation for estimating protein requirements of infants. Because it serves as a guideline for regulatory agencies issuing regulations for infant formula composition, it is critical that information on the protein content of breast milk is reliable. We have therefore carried out a meta-analysis of the protein and amino acid contents of breast milk and how they evolve during lactation. As several bioactive proteins are not completely digested in the infant and therefore represent "non-utilizable" protein, we evaluated the quantity, mechanism of action and digestive fate of several major breast milk proteins. A better knowledge of the development of the protein contents of breast milk and to what extent protein utilization changes with age of the infant will help improve understanding of protein needs in infancy. It is also essential when designing the composition of infant formulas, particularly when the formula uses a "staging" approach in which the composition of the formula is modified in stages to reflect changes in breast milk and changing requirements as the infant ages.
Collapse
Affiliation(s)
- Bo Lönnerdal
- Department of Nutrition, University of California, Davis, USA.
| | - Peter Erdmann
- Nestlé Nutrition, Rue Entre-Deux-Villes 10, CH-1814, La Tour-de-Peilz, Switzerland
| | - Sagar K Thakkar
- Nestlé Research Center, Vers-chez-les-Blanc, P.O. Box 44, CH-1000, Lausanne, 26, Switzerland
| | - Julien Sauser
- Nestlé Research Center, Vers-chez-les-Blanc, P.O. Box 44, CH-1000, Lausanne, 26, Switzerland
| | - Frédéric Destaillats
- Nestlé Nutrition, Rue Entre-Deux-Villes 10, CH-1814, La Tour-de-Peilz, Switzerland
| |
Collapse
|
28
|
Donovan SM. The Role of Lactoferrin in Gastrointestinal and Immune Development and Function: A Preclinical Perspective. J Pediatr 2016; 173 Suppl:S16-28. [PMID: 27234407 DOI: 10.1016/j.jpeds.2016.02.072] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The early postnatal period is a critical time for gastrointestinal (GI) and immune development. Neonates fed mother's milk have more rapid GI and immune development than fed-formula infants. In addition, clinical and epidemiologic data provide strong evidence that breastfeeding reduces the incidence and/or severity of infectious diseases. Lactoferrin is a 77 kDa, iron-binding glycoprotein that is present at high concentration in human milk compared with bovine milk and infant formula. It is a multifunctional protein that mediates many of the physiological processes in which breastfed infants have advantages over their formula-fed peers, including promoting GI and immune development, protection from infections, and improved cognitive development. Feeding bovine lactoferrin or recombinant human lactoferrin was well tolerated and stimulated intestinal cell proliferation and increased villus length and crypt depth in piglets. Lactoferrin also influenced both systemic and GI immune development by stimulating a balanced T-helper-1/T-helper-2 cytokine immune response. Further, there was a tendency for immune cells to secrete more anti-inflammatory cytokines in an unstimulated state, while being primed for a robust pro-inflammatory response when presented with a bacterial trigger in piglets fed lactoferrin. These findings support clinical studies demonstrating benefits of dietary lactoferrin in the prevention of infections, late onset sepsis, and necrotizing enterocolitis.
Collapse
Affiliation(s)
- Sharon M Donovan
- Department of Food Science and Human Nutrition, University of Illinois, Urbana, IL.
| |
Collapse
|
29
|
Guri A, Paligot M, Crèvecoeur S, Piedboeuf B, Claes J, Daube G, Corredig M, Griffiths MW, Delcenserie V. In vitroscreening of mare's milk antimicrobial effect and antiproliverative activity. FEMS Microbiol Lett 2015; 363:fnv234. [DOI: 10.1093/femsle/fnv234] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/03/2015] [Indexed: 12/24/2022] Open
|
30
|
Hoyt AEW, Medico T, Commins SP. Breast Milk and Food Allergy: Connections and Current Recommendations. Pediatr Clin North Am 2015; 62:1493-507. [PMID: 26456446 DOI: 10.1016/j.pcl.2015.07.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Breast milk, a living source of nutrition for babies, complements a baby's immune system, supplementing undeveloped defenses with immune factors while creating the foundation for the innate and adaptive immune systems. Such immune development includes tolerance of the environment and, in the case of food allergy, a lack of tolerance. Recent research questions the previous opinion that breast milk is protective against food allergy. This article reviews the immature immune system, the immunology and nutrition of breast milk, the literature exploring breast milk and food allergy, and the current recommendations regarding breast milk and the prevention of food allergy.
Collapse
Affiliation(s)
- Alice E W Hoyt
- University of Virginia, PO Box 801355, 409 Lane Road MR-4 Building, Room 5051, Charlottesville, VA 22908, USA
| | - Tegan Medico
- University of Virginia, PO Box 801355, 409 Lane Road MR-4 Building, Room 5051, Charlottesville, VA 22908, USA
| | - Scott P Commins
- University of North Carolina School of Medicine, Department of Medicine, Division of Rheumatology, Allergy and Immunology, 3300 Thurston Building, CB 7280, Chapel Hill, NC 27599-7280, USA.
| |
Collapse
|
31
|
Tacoma R, Fields J, Ebenstein DB, Lam YW, Greenwood SL. Characterization of the bovine milk proteome in early-lactation Holstein and Jersey breeds of dairy cows. J Proteomics 2015; 130:200-10. [PMID: 26391770 DOI: 10.1016/j.jprot.2015.09.024] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 09/15/2015] [Accepted: 09/17/2015] [Indexed: 12/25/2022]
Abstract
UNLABELLED Milk is a highly nutritious natural product that provides not only a rich source of amino acids to the consumer but also hundreds of bioactive peptides and proteins known to elicit health-benefitting activities. We investigated the milk protein profile produced by Holstein and Jersey dairy cows maintained under the same diet, management and environmental conditions using proteomic approaches that optimize protein extraction and characterization of the low abundance proteins within the skim milk fraction of bovine milk. In total, 935 low abundance proteins were identified. Gene ontology classified all proteins identified into various cellular localization and function categories. A total of 43 low abundance proteins were differentially expressed between the two dairy breeds. Bioactive proteins involved in host-defense, including lactotransferrin (P=0.0026) and complement C2 protein (P=0.0001), were differentially expressed by the two breeds, whereas others such as osteopontin (P=0.1788) and lactoperoxidase (P=0.2973) were not. This work is the first to outline the protein profile produced by two important breeds of dairy cattle maintained under the same diet, environment and management conditions in order to observe likely true breed differences. This research now allows us to better understand and contrast further research examining the bovine proteome that includes these different breeds. BIOLOGICAL SIGNIFICANCE Within the last decade, the amount of research characterizing the bovine milk proteome has increased due to growing interest in the bioactive proteins that are present in milk. Proteomic analysis of low abundance whey proteins has mainly focused on human breast milk; however, previous research has highlighted the presence of bioactive proteins in bovine milk. Recent publications outlining the cross-reactivity of bovine bioactive proteins on human biological function highlight the need for further investigation into the bovine milk proteome. The rationale behind this study is to characterize and compare the low abundance protein profile in the skim milk fraction produced from Holstein and Jersey breeds of dairy cattle, which are two major dairy cattle breeds in the USA. A combination of fractionation strategies was used to efficiently enrich the low abundance proteins from bovine skim milk for proteomic profiling. A total of 935 low abundance proteins were identified and compared between the two bovine breeds. The results from this study provide insight into breed differences and similarities in the milk proteome profile produced by two breeds of dairy cattle.
Collapse
Affiliation(s)
- Rinske Tacoma
- The University of Vermont, Department of Animal & Veterinary Sciences, 570 Main Street, Burlington, VT 05405, United States
| | - Julia Fields
- The University of Vermont, Vermont Genetics Network Proteomics Facility, 109 Carrigan Drive, Burlington, VT 05405, United States
| | - David B Ebenstein
- The University of Vermont, Department of Animal & Veterinary Sciences, 570 Main Street, Burlington, VT 05405, United States
| | - Ying-Wai Lam
- The University of Vermont, Vermont Genetics Network Proteomics Facility, 109 Carrigan Drive, Burlington, VT 05405, United States
| | - Sabrina L Greenwood
- The University of Vermont, Department of Animal & Veterinary Sciences, 570 Main Street, Burlington, VT 05405, United States.
| |
Collapse
|
32
|
Hwang SA, Kruzel ML, Actor JK. Effects of CHO-expressed recombinant lactoferrins on mouse dendritic cell presentation and function. Innate Immun 2014; 21:553-61. [PMID: 25537452 DOI: 10.1177/1753425914564609] [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: 04/29/2014] [Accepted: 11/15/2014] [Indexed: 12/11/2022] Open
Abstract
Lactoferrin (LF), a natural iron-binding protein, has previously demonstrated effectiveness in enhancing the Bacillus Calmette-Guérin (BCG) tuberculosis vaccine. This report investigates immune modulatory effects of Chinese hamster ovary (CHO) cell-expressed recombinant mouse and human LFs on mouse bone marrow-derived dendritic cells (BMDCs), comparing homologous and heterologous functions. BCG-infected BMDCs were cultured with LF, and examined for class II presentation molecule expression. Culturing of BCG-infected BMDCs with either LF decreased the class II molecule-expressing population. Mouse LF significantly increased the production of IL-12p40, IL-1β and IL-10, while human LF-treated BMDCs increased only IL-1β and IL-10. Overlaying naïve CD4 T-cells onto BCG-infected BMDCs cultured with mouse LF increased IFN-γ, whereas the human LF-exposed group increased IFN-γ and IL-17 from CD4 T cells. Overlay of naïve CD8 T cells onto BCG-infected BMDCs treated with mouse LF increased the production of IFN-γ and IL-17, while similar experiments using human LF only increased IL-17. This report is the first to examine mouse and human recombinant LFs in parallel experiments to assess murine DC function. These results detail the efficacy of the human LF counterpart used in a heterologous system to understand LF-mediated events that confer BCG efficacy against Mycobacterium tuberculosis challenge.
Collapse
Affiliation(s)
- Shen-An Hwang
- Department of Pathology, University of Texas-Houston Medical School, Houston, TX, USA
| | - Marian L Kruzel
- Department of Integrative Biology and Pharmacology, University of Texas-Houston Medical School, Houston, TX, USA
| | - Jeffrey K Actor
- Department of Pathology, University of Texas-Houston Medical School, Houston, TX, USA Program in Immunology, University of Texas Health Science Center, Houston, TX, USA
| |
Collapse
|