Search

Transforming Growth Factor-2 and Epidermal Growth Factors in Medolac Donor Milk and Raw Breast Milk



Breast milk contains critical growth factors for the newborn, especially transforming growth factor (TGF)-b2 and epidermal growth factor (EGF). Ingestion of breast milk TGF-b21-3 and EGF4 may promote gut barrier function, immune tolerance, and mucosal repair in the immature neonatal intestine as well as prevent necrotizing enterocolitis (NEC).


Orally ingested TGF-b2 protected mice5 and baboons6 from NEC by suppressing mucosal inflammatory responses. TGF-b2 was more efficient than TGF-b1 or TGF-b3 in suppressing inflammatory responses in the developing intestine and protecting against NEC.5


Decreased TGF-b2 expression and bioactivity in animal and human intestinal tissues were associated with increased NEC.5 Moreover, ingested EGF could normalize the overproduction of pro-inflammatory cytokines in the neonatal liver with NEC (rat model) by reducing intestinal injury.4


Supplementation of EGF in infant formula decreased the incidence of NEC in a neonatal rat model.7 Ingested EGF also reduced NEC by changing the apoptotic gene and protein expressions in the ileum8 and enhancing the intestinal growth in baby rats.9 Based on those studies, TGF-b2 and EGF in breast milk may protect premature infants against NEC and mucosal injury.


Figure 1: The role of transforming growth factor (TGF)-beta2 and epidermal growth factor (EGF) in the neonatal intestinal development and prevention of NEC.


Objective

The aim of this study was to compare the concentrations of native TGF-b2 and EGF and activated TGF-b2 between 20-Calories Benefit product (retort donor breast milk) and raw breast milk from mothers.


Methods

Concentrations of native and activated TGF-b2 and native EGF were determined via ELISA in three 20-Calories Benefit donor products (Medolac Laboratories) and in raw breast milk samples from sixteen mothers.


For the activation of TGF-b2, the breast milk samples were acidified with 1N HCl to adjust to pH 3 for 15 min, neutralized with 1N NaOH, and then immediately tested. Breast milk supernatants were collected after centrifugation at 3,500 x g for 20 min at 4°C and diluted in 2x with blocking buffer. Primary antibodies (monoclonal mouse anti-human TGF-b2 and EGF), secondary antibodies (polyclonal HRP conjugated rabbit anti-human TGF-b2 and EGF) and purified recombinant TGF-b2 and EGF (for standard curves) were purchased from Aviva System Biology. ELISAs were recorded with a microplate reader SpectraMax iD5 (Molecular Devices) Multiple unpaired t-tests were used for statistical analysis.


Results

Concentrations of native TGF-b2 and activated TGF-b2 in 20-Cal donor milk products were 1.9-(p = 0.02) and 2.6-fold (p = 0.004), respectively, higher than that in raw breast milk (Figure 2). EGF concentration did not differ between 20-Cal donor milk product and raw breast milk (p = 0.64).


Figure 2. Concentrations of native transforming growth factor-b2 (TGF-b2), epidermal growth factor (EGF) and activated TGF-b2 in raw breast milk (n = 16 for mothers) and in 20-Calories Benefit donor breast milk products (n = 3 for batches). Values are mean ± SEM. Asterisks show statistically significant differences between variables (** p < 0.01; * p < 0.05) using multiple unpaired t-tests.

Discussion

Several active breast milk components with immune-protective functions are affected during pasteurization, including lactoferrin (57–80% reduction), secretory IgA (22–48% reduction), IgM (98–100% reduction) and IgG (34–41% reduction),10-12 therefore retort processing will also decrease their concentrations. However, TGF-b and EGF were not affected by pasteurization.13,14 No previous study has determined whether TGF-b2 and EGF are affected during the retort processing.


For the first time, we found that native and activated TGF-b2 concentrations in Medolac 20-Cal donor milk breast products were higher than in raw breast milk samples. This increase of concentration in TGF-b2 could be due to the heat-treatment during retort processing and the mixture of raw human whole milk and cream. These results are in agreement with Namachivayam et al. (2013)15 that TGF-b2 bioactivity in both pasteurized donor breast milk and raw breast milk (preterm-delivering at 1 week and 1 month of postpartum) was increased after heat-treatment at 80°C for 5 min. These authors used this heat-treatment to activate latent TGF-b2. Moreover, Reeves et al.16 observed that TGF-b2 concentration in pasteurized donor breast milk fortifier (HDMF) was higher than pasteurized donor breast milk due to its processing; HDMF is made from pooled human milk creams that may concentrate TGF-b2.


Latent TGF-b in breast milk is activated to the immunoreactive isoform by acidification (pH 3), which denatures latency-associated peptide (LAP), a peptide binds to TGF-b.17 Other mechanisms can activate TGF-b, such as proteases and heat-treatment.18,19 Pre-activation of breast milk TGF-b using heat-treatment or acidification/neutralization is particularly critical for premature infants due to their higher gastric pH and lower protease activity in the stomach and intestine compared with term infants.20 The capacity to activate TGF-b during digestion in preterm infants is unknown and future studies are needed to understand the functional activity of TGF-b in the neonatal gut.


Conclusions

Medolac 20-Cal donor breast milk products contain higher TGF-b2 concentration compared with raw breast milk due to the activation of latent TGF-b2 by the retort heat-treatment and concentrated TGF-b2 in product formulation. Higher levels of TGF-b2 in Medolac donor breast milk products may improve the protection of preterm infants against NEC/mucosal injury and enhance the maturation of the gastrointestinal tract.



References


1. Oddy, W.H.a.M., R.J. Milk‐derived or recombinant transforming growth factor‐beta has effects on immunological outcomes: a review of evidence from animal experimental studies. Clinical & Experimental Allergy 2011, 41, 783-793.

2. Rautava, S., Lu, L., Nanthakumar, N.N., Dubert-Ferrandon, A. and Walker, W.A.,. TGF-β2 induces maturation of immature human intestinal epithelial cells and inhibits inflammatory cytokine responses induced via the NF-κB pathway. J Pediatr Gastroenterol Nutr 2012, 54, 630-638.

3. Rautava, S., Nanthakumar, N.N., Dubert-Ferrandon, A., Lu, L., Rautava, J. and Walker, W.A. Breast milk-transforming growth factor-β2 specifically attenuates IL-1β-induced inflammatory responses in the immature human intestine via an SMAD6-and ERK-dependent mechanism. Neonatology 2011, 99, 192-201.

4. Halpern, M.D., Holubec, H., Clark, J.A., Saunders, T.A., Williams, C.S., Dvorak, K. and Dvorak, B. Epidermal growth factor reduces hepatic sequelae in experimental necrotizing enterocolitis. Neonatology 2006, 89, 227.

5. Maheshwari, A., Kelly, D.R., Nicola, T., Ambalavanan, N., Jain, S.K., Murphy–Ullrich, J. et al. TGF-β2 suppresses macrophage cytokine production and mucosal inflammatory responses in the developing intestine. Gastroenterology 2011, 140, 242-253.

6. Namachivayam, K., Blanco, C.L., MohanKumar, K., Jagadeeswaran, R., Vasquez, M., McGill-Vargas, L., et al. Smad7 inhibits autocrine expression of TGF-β2 in intestinal epithelial cells in baboon necrotizing enterocolitis. American Journal of Physiology-Gastrointestinal and Liver Physiology, 2012, 304, G167-G180.

7. Dvorak, B., Halpern, M.D., Holubec, H., Williams, C.S., McWilliam, D.L., Dominguez, J.A., et al. Epidermal growth factor reduces the development of necrotizing enterocolitis in a neonatal rat model. American Journal of Physiology-Gastrointestinal and Liver Physiology, 2002, 282, G156-G164.

8. Clark, J.A., Lane, R.H., MacLennan, N.K., Holubec, H., Dvorakova, K., Halpern, M.D., et al. Epidermal growth factor reduces intestinal apoptosis in an experimental model of necrotizing enterocolitis. American Journal of Physiology-Gastrointestinal and Liver Physiology 2005, 288, G755-G762., doi:10.1152/ajpgi.00172.2004.

9. Berseth, C.L. Enhancement of intestinal growth in neonatal rats by epidermal growth factor in milk. American Journal of Physiology-Gastrointestinal and Liver Physiology 1987, 253, G662-G665.

10. Czank, C., Prime, D.K., Hartmann, B., Simmer, K. and Hartmann, P.E. Retention of the immunological proteins of pasteurized human milk in relation to pasteurizer design and practice. Pediatric research 2009, 66, 374.

11. Ford, J.; Law, B.; Marshall, V.M.; Reiter, B. Influence of the heat treatment of human milk on some of its protective constituents. J Pediatr 1977, 90, 29-35.

12. Demers-Mathieu, V.; Huston, R.K.; Markell, A.M.; McCulley, E.A.; Martin, R.L.; Spooner, M.; Dallas, D.C. Differences in Maternal Immunoglobulins within Mother’s Own Breast Milk and Donor Breast Milk and across Digestion in Preterm Infants. Nutrients 2019, 11, 920.

13. Goelz, R., Hihn, E., Hamprecht, K., Dietz, K., Jahn, G., Poets, C. and Elmlinger, M. Effects of different CMV-heat-inactivation-methods on growth factors in human breast milk. Pediatric research 2009, 65, 458.

14. McPherson, R.J.a.W., C.L. The effect of pasteurization on transforming growth factor alpha and transforming growth factor beta 2 concentrations in human milk. In In Bioactive Components of Human Milk, Springer, B., MA., Ed. 2001; pp. 559-566.

15. Namachivayam, K., Blanco, C.L., Frost, B.L., Reeves, A.A., Jagadeeswaran, R., MohanKumar, K., et al. Preterm human milk contains a large pool of latent TGF-β, which can be activated by exogenous neuraminidase. . American Journal of Physiology-Gastrointestinal and Liver Physiology 2013, 304, G1055-G1065.

16. Reeves, A.A., Johnson, M.C., Vasquez, M.M., Maheshwari, A. and Blanco, C.L. TGF-β2, a protective intestinal cytokine, is abundant in maternal human milk and human-derived fortifiers but not in donor human milk. Breastfeeding Medicine 2013, 8, .496-502.

17. Rigotti, E., Piacentini, G.L., Ress, M., Pigozzi, R., Boner, A.L. and Peroni, D.G. Transforming growth factor‐β1 and interleukin‐10 in breast milk and development of atopic diseases in infants. Clinical & Experimental Allergy 2006, 36, 614-618.

18. Nakamura, Y., Miyata, M., Ando, T., Shimokawa, N., Ohnuma, Y., Katoh, R., et al. The latent form of transforming growth factor-β administered orally is activated by gastric acid in mice. . The Journal of nutrition 2009, 139, 1463-1468.

19. Jenkins, G. The role of proteases in transforming growth factor-β activation. . The international journal of biochemistry & cell biology 2008, 40, 1068-1078.

20. Demers-Mathieu, V.; Qu, Y.; Underwood, M.A.; Borghese, R.; Dallas, D.C. Premature infants have lower gastric digestion capacity for human milk proteins than term infants. Journal of pediatric gastroenterology and nutrition 2018, 66, 816-821.



About the Author

Dr. Veronique Demers-Mathieu is the Senior Research Scientist in the department of Neonatal Immunology and Microbiology at Medolac Laboratories. She did 3-years of postdoctoral training in Neonatal Nutrition under Dr. Dallas at Oregon State University and earned her Ph.D. in Food Microbiology at Laval University. Her expertise focuses on the immune components (including antibodies, immune cells, cytokines, and bioactive proteins) from human milk that protect infants against infectious diseases.