Hostname: page-component-8448b6f56d-dnltx Total loading time: 0 Render date: 2024-04-23T06:08:25.932Z Has data issue: false hasContentIssue false

Non-nutritive bioactive components in maternal milk and offspring development: a scoping review

Published online by Cambridge University Press:  07 April 2022

Shafinaz Eisha
Affiliation:
Department of Biological Sciences, Center for Environmental Epigenetics and Development, University of Toronto Scarborough, Toronto, ON, Canada Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
Ishraq Joarder
Affiliation:
Department of Biological Sciences, Center for Environmental Epigenetics and Development, University of Toronto Scarborough, Toronto, ON, Canada
Sanoji Wijenayake
Affiliation:
Department of Biological Sciences, Center for Environmental Epigenetics and Development, University of Toronto Scarborough, Toronto, ON, Canada Department of Biology, Richardson College for the Environment and Science Complex, The University of Winnipeg, Winnipeg, MB, Canada
Patrick O. McGowan*
Affiliation:
Department of Biological Sciences, Center for Environmental Epigenetics and Development, University of Toronto Scarborough, Toronto, ON, Canada Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada Department of Psychology, University of Toronto, Toronto, ON, Canada Department of Physiology, University of Toronto, Toronto, ON, Canada
*
Address for correspondence: Patrick O. McGowan, Ph.D., University of Toronto, Scarborough Campus, 1265 Military Trail, Toronto, Ontario, Canada, M1C1A4. E-mail: patrick.mcgowan@utoronto.ca
Rights & Permissions [Opens in a new window]

Abstract

Lactation is a critical time in mammalian development, where maternal factors shape offspring outcomes. In this scoping review, we discuss current literature concerning maternal factors that influence lactation biology and highlight important associations between changes in milk composition and offspring outcomes. Specifically, we explore maternal nutritional, psychosocial, and environmental exposures that influence non-nutritive bioactive components in milk and their links to offspring growth, development, metabolic, and behavioral outcomes. A comprehensive literature search was conducted using the Preferred Reporting Items for Systematic Reviews and Meta-Analysis Extension for Scoping Reviews (PRISMA-ScR) guidelines. Predetermined eligibility criteria were used to analyze 3,275 papers, and the final review included 40 primary research articles. Outcomes of this review identify maternal obesity to be a leading maternal factor influencing the non-nutritive bioactive composition of milk with notable links to offspring outcomes. Offspring growth and development are the most common modes of programming associated with changes in non-nutritive milk composition due to maternal factors in early life. In addition to discussing studies investigating these key associations, we also identify knowledge gaps in the current literature and suggest opportunities and considerations for future studies.

Type
Review
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an Open Access article, distributed under the terms of the Creative Commons Attribution licence (https://creativecommons.org/licenses/by/4.0/), which permits unrestricted re-use, distribution, and reproduction in any medium, provided the original work is properly cited.
Copyright
© The Author(s), 2022. Published by Cambridge University Press in association with International Society for Developmental Origins of Health and Disease

Background

Maternal milk is a heterogeneous biological fluid that is tailored to meet the developmental, digestive, and immune needs of offspring and is the sole source of nutrition for most newborn mammals. Reference Andreas, Kampmann and Mehring Le-Doare1Reference Beghetti, Biagi, Martini, Brigidi, Corvaglia and Aceti3 The World Health Organization (WHO) recommends that infants should be exclusively breastfed for a minimum of 6 months to receive the optimal benefits of maternal milk. Reference Kramer and Kakuma4 Milk is rich in nutritive components, including carbohydrates, proteins, lipids, vitamins, and minerals, which have been discussed in relation to offspring development elsewhere. Reference Andreas, Kampmann and Mehring Le-Doare1,Reference Ballard and Morrow2,Reference Allen5Reference Stannard, Miller and Old10 Milk also contains non-nutritive bioactive components, including growth and neurodevelopmental factors, hormones, active enzymes, peptides, a complex microbiota, immune factors, as well as non-coding RNA (microRNA – miRNA) encapsulated in milk-derived exosomes, which play roles in immune maturation, metabolism, overall health and developmental outcomes of offspring. Reference Zamanillo, Sánchez, Serra and Palou11Reference Zempleni, Aguilar-Lozano and Sadri18

Maternal milk composition varies across lactation stages, milk fractions, circadian cycle, and even temporally within a single feed, exhibiting compositional differences between foremilk and hindmilk. Reference Andreas, Kampmann and Mehring Le-Doare1,Reference Ballard and Morrow2,Reference McGuire, Seppo and Goga19,Reference Bernstein and Hinde20 The first fluid produced by mammary gland epithelial cells soon after delivery, known as colostrum, is rich in non-nutritive factors, including hormones, growth factors and immune components (e.g., lactoferrin, leukocytes, and immunoglobulins). Reference Ballard and Morrow2,Reference Keikha, Shayan-Moghadam, Bahreynian and Kelishadi8,Reference Puppel, Gołębiewski and Grodkowski21 In humans, colostrum is replaced by transition milk approximately 60 hours to 5 days post birth and lasts for about 10 days to 2 weeks post-partum, after which mature milk production is initiated. Reference Ballard and Morrow2,Reference Boss, Gardner and Hartmann22,Reference Christian, Smith, Lee, Vargas, Bremer and Raiten23

Studies in human and animal models suggest that non-nutritive components in maternal milk contribute to early life developmental programming. Developmental programming refers to cellular and biochemical processes that lead to long-term functional changes as a result of a stimulus and/or insult during a critical period of development. Reference Langley-Evans24 For example, maternal nutrition and psychosocial stress exposure during prenatal and/or early postnatal period can affect the overall growth and cognitive development of offspring, as well as increase the risk of disease later in life. Reference Alfaradhi and Ozanne25Reference Aizer, Stroud and Buka27 Non-nutritive bioactive compounds in milk enhance offspring survival by promoting the healthy development of the immune system and intestinal microbial colonization. Reference Ballard and Morrow2,Reference Le, Holder, Bassett and Pannaraj14,Reference Oftedal28Reference Azad, Konya and Maughan31 Emerging evidence also suggests that these bioactive components in milk may help protect infants against acute infections, neuroinflammation and systemic inflammation, and metabolic diseases, including obesity, hypertension, and/or type I and II diabetes. Reference Andreas, Kampmann and Mehring Le-Doare1,Reference Ballard and Morrow2,Reference Munblit, Treneva and Peroni12,Reference Badillo-Suárez, Rodríguez-Cruz and Nieves-Morales32,Reference Mirza, Kaur and Nielsen33

There is considerable evidence that perinatal (e.g., the combined period of prenatal and postnatal life) maternal overnutrition and psychosocial stress play a role in shaping metabolic and neurodevelopmental outcomes in the offspring. Reference Chen, Wang and Yang34,Reference Fodor and Zelena35 For example, in rodent models, a maternal diet high in saturated fats and complex sugars leads to increased offspring bodyweight, Reference Chen, Wang and Yang34 elevated levels of inflammation in visceral adipocytes, Reference Monks, Orlicky and Stefanski36 increased liver steatosis in males, Reference Monks, Orlicky and Stefanski36 and increased anxiety-like behavior that persists into adulthood. Reference Sasaki, de Vega, St-Cyr, Pan and McGowan37,Reference Winther, Elfving, Müller, Lund and Wegener38 In humans, maternal obesity is associated with an increased risk of childhood asthma, and higher infant weight-for-length and Body Mass Index (BMI) at birth. Reference Ellsworth, Perng, Harman, Das, Pennathur and Gregg39,Reference Forno, Young, Kumar, Simhan and Celedón40 However, the milk of mothers with obesity was found to contain high levels of insulin and leptin, which may provide some benefit to the developing offspring, Reference Chan, Goruk and Becker41 as increased levels of milk insulin and leptin reduce intestinal inflammation and increase intestinal barrier function. Reference Lemas, Young and Baker42

Maternal anxiety during gestation as well as psychological and social stress are associated with a slower rate of cognitive development (lower scores on the mental development index of the Bayley Scales of Infant Development) in human infants over the first postnatal year. Reference Davis and Sandman43 In addition, maternal psychosocial stress is correlated with lower levels of milk bacterial diversity at 3 months post-partum, Reference Browne, Aparicio and Alba44 which may contribute to a subsequent decrease in infant gut microbiome diversity. Reference Pannaraj, Li and Cerini45 While still debated, Reference Carlson, Xia and Azcarate-Peril46 there is evidence that this decreased complexity of the neonatal gut microbiome is associated with elevated risks of gastrointestinal, autoimmune, and metabolic disorders in adulthood. Reference Milani, Duranti and Bottacini47,Reference Turroni, Milani and Duranti48 Although the compositional changes of non-nutritive components in milk due to maternal obesity and nutritional status is well-established, Reference Bautista, Montaño and Ramirez49Reference Pomar, Castro, Picó, Serra, Palou and Sánchez57 less is known about how maternal psychosocial stress during the pre-gestational/gestational/post-gestational periods impact milk composition. Reference Thibeau, D’Apolito and Minnick58,Reference Chen, Nommsen-Rivers, Dewey and Lönnerdal59 Also, while a majority of studies with human subjects has statistically controlled for anthropometric and demographic covariates, including maternal height, age and ethnicity, these measurements have not been investigated as independent variables that could potentially alter non-nutritive milk composition. This is particularly important because milk composition varies across demographics. For instance, maternal age and ethnicity are highly correlated with fat content in milk. Reference Denić, Sunarić and Genčić60,Reference Butts, Hedderley and Herath61

This scoping review was designed to provide a comprehensive overview of studies that have investigated non-nutritive bioactive components in milk associated with maternal factors and/or physiological and behavioral outcomes of offspring. In addition to a focus on the human literature, we did not exclude model organisms during the screening process to avoid missing relevant literature that fit within the scope of our study. Model systems that have been investigated in lactation biology research are highly heterogenous and span non-human primates, rodents, bovine, and other mammals. We categorized the primary literature into different species to aid in elucidating knowledge gaps in lactation biology. We attempted to characterize emerging knowledge concerning the most studied maternal factors that play roles in altering the non-nutritive bioactive components of maternal milk, along with their associations with offspring outcomes during early childhood that can potentially persist into adulthood.

Methods

This scoping review was performed in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analysis Extension for Scoping Reviews (PRISMA-ScR) guidelines. Reference Tricco, Lillie and Zarin62

Search strategy

A comprehensive literature search was performed using the databases MEDLINE, PubMed, EMBASE and the Web of Science, with “and/or” combinations of the following keywords: mammalian milk, lactation biology, maternal factors, and offspring developmental programming. The literature search was last updated on August 5th, 2020. All records were imported into Covidence systematic review software (Veritas Health Innovation, Melbourne, Australia) and duplicates were removed from the library.

Exclusion/eligibility criteria and article selection

Title and abstract screening were conducted by two, independent reviewers (IJ and SE), with disagreements resolved by a third independent reviewer (SW). During the abstract and title screening phase, articles were excluded if one or more of the following criteria were met: 1) Primary language is not English; 2) Not peer-reviewed; 3) Sole focus on metabolic, nutritional, and immune stress affecting maternal health as opposed to mother-offspring dyads or maternal factors contributing to changes in milk composition; 4) Do not link changes in milk composition with either maternal factors or offspring outcomes.

Remaining articles were subcategorized into four species-specific databases (human, rodent, bovine, and other mammals). We included studies that spanned human and non-human models, because many previously published studies used several non-human mammalian model systems to investigate different aspects of lactation biology that were relevant to our research objective. For instance, numerous studies have investigated the mechanistic and molecular mechanisms of milk derived exosome uptake and transfer using rodent, bovine, and marsupial milk. However, studies that have investigated non-nutritive milk components in relation to maternal factors and/or offspring outcomes mainly used human and/or rodent models. Further, differences in milk composition have been reported across mammals. Reference Edwards, Lavergne and McCaw63 As such, we decided to include all relevant studies rather than to exclude non-human model systems, and used four subcategories to differentiate human studies from studies of rodents, bovines, and other mammals.

Subsequently, the remaining articles were subjected to full-text screening. As per abstract and title screening, two independent reviewers (IJ and SE) conducted this analysis, and a third independent reviewer (SW) resolved disagreements. The following criteria were used to determine eligibility of the full-text articles towards the scoping review. Studies that contained the following criteria were included: 1) Primary literature; 2) A primary focus on non-nutritive bioactive components of milk as opposed to nutritive components; 3) Report experimentally validated physiological and psychological outcomes in offspring as opposed to conducting only in vitro experiments and/or in-silico predictions; 4) Research topics investigating non-nutritive bioactive components in milk and relating them to maternal factors and/or offspring outcomes (Table 1); 5) Experimental methodologies for milk exosome isolation and characterization adhering to the guidelines put forth by the International Society for Extracellular Vesicles (ISEV), where applicable. Reference Théry, Witwer and Aikawa64

Table 1. Search terms and strategy

Note: We searched for primary literature that included non-nutritive bioactive components in maternal milk AND linked that to maternal factors AND/OR offspring outcomes. We used multiple search terms (individually and in combination) for each category, and we included papers that contained at least one of the search terms.

Data collection and extraction

Data extraction was conducted for all articles that remained post full-text screening. A comprehensive and detailed excel inventory was created as a data charting form and the extractable variables were jointly developed by three reviewers (IJ, SE and SW). Extracted data included study characteristics (author, year of publication, country and doi), subtopic categorization based on overarching research objectives, maternal factors in relation to non-nutritive lactation biology and developmental outcomes of offspring. Two reviewers (IJ and SE) independently extracted and inputted data based on the distinct categorical variables developed.

Results

Study selection

A total of 4,741 journal articles were identified after the primary search. One thousand four hundred and sixty-six duplicates were removed using the Covidence reference manager and the 3,275 papers that remained were subjected to first round eligibility screening. Upon completion, 233 articles met the first eligibility criteria and were organized into four databases based on the species that was investigated, where the human database contained 142 entries, rodent contained 57, bovine contained 37, and the other category involving all other mammals contained 24. Fifteen papers were repeated across multiple species at this stage of screening. Finally, 40 articles passed the second round of full-text eligibility screening and were used in the final review (Fig. 1). The final collection of 40 papers did not have repeating entries.

Fig. 1. PRISMA-ScR flow chart illustrating screening and eligibility criteria for article selection.

Study characteristics and categorization

The final selection of 40 primary research articles that remained post-screening and eligibility testing were categorized into two overarching research objectives, with a third category that linked both objectives, across four different species (Table 2). Fourteen papers (35.0%) pertained to maternal factors influencing non-nutritive bioactive components in milk, 13 papers (32.5%) related to non-nutritive components of milk impacting offspring development, and 13 papers (32.5%) linked maternal factors altering non-nutritive bioactive milk components and, in turn, alterations in offspring outcomes. Of the 40 papers, 25 papers (62.5%) were in humans, 8 papers (20.0%) were in rodents, 2 papers (5.0%) in bovine and 5 papers (12.5%) were in other mammals.

Table 2. Overarching research topics and categorization of the articles included in the final review (N = 40)

Analysis and synthesis of results

Of the 40 remaining papers, 13 papers (32.5%) characterized non-nutritive bioactive components in milk in relation to maternal factors impacting developmental outcomes of offspring (Table 2 – Category 3; Table 3). Within this category, six papers (46.1%) were in human, five papers (38.5%) were in rodent, and two papers (15.4%) were in other mammals, specifically rhesus macaques (Macaca mulatta). As reported in Table 3, 11 studies (84.6%) investigated maternal overweight or obesity status, which was associated with altering non-nutritive bioactive milk components and impacting offspring outcomes. Maternal gestational stress, metabolic syndrome (gestational diabetes mellitus), social rank, and parity were investigated by only four studies (30.1%). Two of these studies were included in multiple categories since they investigated more than one maternal factor (e.g., obesity, gestational stress and metabolic syndrome). In addition, weight, growth, and development were the most common offspring outcomes that were explored across all 13 papers with respect to maternal factors and non-nutritive milk composition. Five papers (38.4%) measured offspring body fat/adiposity, four papers (30.8%) pertained to offspring metabolism, and two papers (15.4%) examined offspring behavior. Finally, two papers (15.4%) assessed other offspring outcomes, such as microbiome, intestinal, and adipose tissue inflammation.

Table 3. Non-nutritive bioactive components in milk linking maternal factors with offspring outcomes

1–13Numerical values in “Offspring outcomes” correspond to the references listed in the “Citations” column.

a Characterization of obesity in human studies: overweight: BMI > 25 kg/m2, and obese: BMI > 30 kg/m2.

b Growth: Z-scores for weight-for-age, length-for-age, head circumference-for-age, weight-for-length, body muscle index.

c Development: brain, immunity.

d Behavior: nervous and confident temperament, milk ingestion.

Supplementary Table 1 contains a complete list of all 40 papers identified in the final selection of primary literature included in this scoping review, along with a short description of each paper’s main findings/focus.

Discussion

The overall objective of this scoping review was to explore the current literature pertaining to maternal factors, including nutritional status, psychosocial stress, and environmental factors that may influence non-nutritive bioactive composition in milk, and thereby impact developmental trajectories in offspring (e.g., physiological, neurological, metabolic, and behavioral outcomes). Numerous studies have investigated distinct aspects of this topic and the associations between individual maternal factors, lactation biology, and/or offspring outcomes. However, potential causal relationships and associations between maternal factors, changes in non-nutritive milk composition, and early life programming effects in offspring remain poorly understood.

Maternal factors influencing milk composition and offspring developmental trajectories

Maternal factors, including nutritional status, metabolic disorders, and/or psychosocial stress appear to play important roles in shaping non-nutritive milk composition (explored across 14 papers; Table 2), which in turn may influence the development and early life programming of offspring (explored across 13 papers; Table 2). Maternal nutritional status was the most investigated maternal factor reported to influence lactation biology and offspring outcomes. In particular, there is evidence that maternal obesity status (BMI > 30 kg/m2) alters miRNAs, Reference Xi, Jiang, Li, Chen, Song and Li65 immune components, Reference Fujimori, França, Morais, Fiorin, de Abreu and Honório-França66 pro-inflammatory fatty acid, Reference Panagos, Vishwanathan and Penfield-Cyr50 and carotenoid levels Reference Panagos, Vishwanathan and Penfield-Cyr50 in milk. These bioactive compounds are critical for offspring growth Reference Ellsworth, Perng, Harman, Das, Pennathur and Gregg39 , visual and neural development, Reference Panagos, Vishwanathan and Penfield-Cyr50 intestinal microbiota diversity and metabolic health. Reference Lemas, Young and Baker42 Moreover, human and laboratory rodent studies have demonstrated that the initiation of lactation was delayed in mothers that were on a high fat diet, Reference Hernandez, Grayson, Yadav, Seeley and Horseman67,Reference Liu, Smith, Dobre and Ferguson68 including disruption of mammary gland morphology and a notable decline of intact alveolar units that are essential for lactogenesis. Reference Hernandez, Grayson, Yadav, Seeley and Horseman67 Several rodent studies also reported that mothers exposed to a high fat diet during pre-gestation, gestation, and lactation, exhibited increased nursing behaviors, where time spent in “arched-back” Reference Purcell, Sun, Pass, Power, Moran and Tamashiro69 and “blanket” Reference Abuaish, Spinieli and McGowan70 nursing increased during the dark phase of the circadian cycle. “Arched-back” nursing is a type of maternal care behavior, where the mother arches her back while nursing pups to allow more access to the teats, and “blanket” nursing is where the dam uses her body to lay over the pups while nursing. Reference Champagne, Diorio, Sharma and Meaney71,Reference Myers, Brunelli, Squire, Shindeldecker and Hofer72 We previously postulated that the percent increase in time spent nursing may be a compensatory lactation-specific behavior to combat impaired milk production, prolactin insensitivity Reference Buonfiglio, Ramos-Lobo and Freitas73 or delayed milk production due to increased mammary gland inflammation. Reference Hernandez, Grayson, Yadav, Seeley and Horseman67,Reference Abuaish, Spinieli and McGowan70 Several human studies have shown that maternal obesity alters hormonal and immune components in milk, and that being obese and/or overweight during the pre-gestational period is associated with increased bodyweight and alterations in the metabolic profile of offspring during early childhood. Reference Ellsworth, Perng, Harman, Das, Pennathur and Gregg39,Reference Larnkjær, Ong, Carlsen, Ejlerskov, Mølgaard and Michaelsen74,Reference Lagström, Rautava and Ollila75 For example, higher maternal post-pregnancy BMI was associated with increased levels of milk adiponectin, a hormone that affects development by regulating lipid metabolism, reducing pro-inflammatory cytokines and improving insulin sensitivity in offspring. Reference Martin, Woo and Geraghty76Reference Yu, Rong and Sun79 Other studies reported that adiponectin levels in maternal milk from healthy lactating women were found to be positively correlated with adiponectin in infant serum Reference Savino, Lupica, Benetti, Petrucci, Liguori and Cordero Di Montezemolo80 and inversely associated with growth and weight gain during early infancy. Reference Yu, Rong and Sun79,Reference van Rossem, Smit and Lentjes81 Another study reported that infants who were partially breastfed (defined as the combined use of maternal milk and formula) by mothers with obesity had elevated levels of insulin and adiponectin and lower levels of IGF-I (insulin growth factor 1) in the blood at 9 months of age compared to infants partially breastfed by normal weight mothers. Reference Larnkjær, Ong, Carlsen, Ejlerskov, Mølgaard and Michaelsen74 Thus, the higher concentration of blood adiponectin reported in offspring who were born to as well as partially breastfed by mothers with obesity may indicate a positive, lactation-specific metabolic adaptation to prenatal maternal obesity exposure.

Maternal psychosocial stress is another prominent factor that may influence cortisol levels in milk, Reference Aparicio, Browne and Hechler82 which in turn has been associated with offspring development and behavioral trajectories. A study in rhesus macaques reported a positive correlation between increased maternal milk cortisol levels and changes in male offspring temperament, including increased activity, boldness, confidence and curiosity. Reference Sullivan, Hinde, Mendoza and Capitanio83 Higher milk cortisol was also associated with increased social interactions in female macaques at 4–8 months of age. Reference Dettmer, Murphy and Guitarra84 However, another study in rhesus macaques found that cortisol levels in milk were positively correlated with nervous temperament, and negatively correlated with confident temperament in offspring. Reference Hinde, Skibiel, Foster, Del Rosso, Mendoza and Capitanio85 A study in humans reported a protective effect of increased milk cortisol exposure during early childhood, where increased milk cortisol levels were associated with decreased childhood obesity. Reference Hahn-Holbrook, Le, Chung, Davis and Glynn86 However, there is other evidence that higher milk cortisol levels are associated with increased fear reactivity in female infants at 3 months Reference Grey, Davis, Sandman and Glynn87 and 8 months of age. Reference Nolvi, Uusitupa and Bridgett88 Despite mixed findings on the beneficial/deleterious effects of increased milk cortisol transfer across mother-offspring dyads, these studies illustrate the manner in which maternal psychosocial stress may alter milk composition and offspring behavior.

Additional studies in humans and other mammals reported that maternal immune status, group rank, and parity also influence not only cortisol, but adiponectin, growth factors and secretory immunoglobulin levels in milk. Reference Sullivan, Hinde, Mendoza and Capitanio83,Reference Groer, Davis and Steele89,Reference Bunney, Zink, Holm, Billington and Kotz90 Gestational age and mode of delivery are two other maternal factors associated with altered milk composition. Reference Carney, Tarasiuk and Diangelo91 Specifically, Carney et al. Reference Carney, Tarasiuk and Diangelo91 reported that the expression profiles of milk miRNAs in lipid and skim fractions differed in preterm delivery compared to term birth. They found that two miRNAs showed increased transcript abundance, and six miRNAs showed decreased transcript abundance in both fractions of preterm milk, when compared to term milk. In addition, six miRNAs in maternal milk were correlated (three positively and three negatively) with mode of delivery. Reference Carney, Tarasiuk and Diangelo91 None of the expression profiles of the milk miRNAs examined in the study were associated with maternal age, race, or ethnicity. Reference Carney, Tarasiuk and Diangelo91

Limitations in the current literature

We identified several common limitations in studies of human lactation to date. First, limited number of studies included large heterogeneous populations representing a wide range of ethnicities, socioeconomic status, and pre-gestational and gestational BMIs. Although it may be challenging to include multi-ethnic populations due to limitations in funding as well as difficulties in the recruitment of mothers belonging to underrepresented groups, the lack of diversity in these datasets limits the generalizability of the interpretations relating to lactation biology and offspring development. Interestingly, the few studies that included diverse populations reported differences in milk composition among ethnic groups as a function of regional maternal diets and breastfeeding practices. Reference Gay, Koleva and Slupsky92,Reference Su, Thamarai Chelvi and Lim93 Second, a large proportion of studies lack detailed descriptions of the methods used for milk collection and analysis, including the time of milk collection, use of breast pumps, the interval from last feeding to the time of sample collection, storage and processing steps, and lactational age. Consideration of these factors is critically important for studies that use human milk, especially human mature milk since there are notable differences in bioactive composition as mature milk is produced over a longer period of time, ranging from 4 to 6 weeks post-partum until the end of the lactation period. Reference Ballard and Morrow2 In addition, milk collected at the beginning of a feed (foremilk) contains lower fat and higher cellular content compared to the milk collected at the end of the feed (hindmilk). Reference McGuire, Seppo and Goga19 Efforts to standardize the methods, protocols, techniques for milk sample collection and processing by following the technical guidelines set forth by the ISEV (referred to as the Minimal Information for Studies of Extracellular Vesicles, MISEV) could serve to limit these potential confounds. Reference Théry, Witwer and Aikawa64

Limitations of this scoping review

While we have attempted to provide a comprehensive analysis of the current literature on the biological associations between maternal factors, lactation biology and developmental outcomes in offspring, this scoping review is not without limitations. Despite compiling 40 papers from four different databases, we may have missed relevant studies in our search. Only peer-reviewed, primary literature published in English was included in our search criteria. A few published studies required specialty permission and journal subscriptions that limited our ability to access the full-print version to apply the exclusion and inclusion criteria. Moreover, the anthropometric controls, such as maternal obesity, used as fixed variables in epidemiological studies varied widely, and many studies investigated only one or two maternal factors at distinct timeframes. For instance, while some studies examined maternal pre-gestational obesity, Reference Ellsworth, Perng, Harman, Das, Pennathur and Gregg39,Reference Lemas, Young and Baker42,Reference Panagos, Vishwanathan and Penfield-Cyr50 others investigated maternal gestational or post-gestational obesity. Reference Zamanillo, Sánchez, Serra and Palou11,Reference Larnkjær, Ong, Carlsen, Ejlerskov, Mølgaard and Michaelsen74 This imposed limitations on our ability to draw clear conclusions from such findings concerning the roles of specific factors.

Conclusion

There is strong evidence that maternal factors, including nutritional status, psychosocial stress, and environmental factors, influence the non-nutritive bioactive components in milk, which are associated with developmental trajectories in offspring. These outcomes include early life growth, weight, height, language, and emotional development. Studies using non-human models have provided important information concerning the mechanistic role of non-nutritive bioactive components of milk on offspring development. Understanding the association between milk composition and offspring developmental outcomes is essential in the development of strategies to enhance maternal nutrition and mitigate risk factors, leading to the improved health of both the mother and offspring. Milk compositional analysis may also inform enhancement of the quality of infant formula, given that exclusive breastfeeding is not feasible for all mothers due to health and/or lactational complications. Future studies with an increased ethnic and socioeconomic diversity of participants, a more extensive collection of participant phenotypes, and more standardized procedures for human milk collection, processing, storage, and analysis will enhance the generalizability and reproducibility of knowledge in this emerging field of research.

Supplementary materials

For supplementary material for this article, please visit https://doi.org/10.1017/S2040174422000149

Acknowledgements

We would like to thank Ms. Sara Guay, Liaison Librarian at University of Toronto – Scarborough for assisting with the literature search. The authors are entirely responsible for the scientific content of the paper.

Financial support

This work was supported by the Natural Sciences and Engineering Council of Canada (NSERC) (P.O.M., grant number RGPIN-2019-493091), (I.J., Undergraduate Student Research Award), and (S.W., Post-doctoral Research Fellowship).

Conflicts of interest

The authors declare that they have no competing or financial interests.

Ethical standards

None.

Footnotes

Shafinaz Eisha and Ishraq Joarder contributed equally to the manuscript.

References

Andreas, NJ, Kampmann, B, Mehring Le-Doare, K. Human breast milk: a review on its composition and bioactivity. Early Hum Dev. 2015; 91(11), 629635. DOI 10.1016/j.earlhumdev.2015.08.013.Google ScholarPubMed
Ballard, O, Morrow, AL. Human milk composition. Nutrients and bioactive factors. Pediatr Clin North Am. 2013; 60(1), 4974. DOI 10.1016/j.pcl.2012.10.002.CrossRefGoogle ScholarPubMed
Beghetti, I, Biagi, E, Martini, S, Brigidi, P, Corvaglia, L, Aceti, A. Human milk’s hidden gift: implications of the milk microbiome for preterm infants’ health. Nutrients. 2019; 11(12), 2944. DOI 10.3390/nu11122944.Google ScholarPubMed
Kramer, MS, Kakuma, R. The optimal duration of exclusive breastfeeding: a systematic review. Adv Exp Med Biol. 2004; 554, 6377. DOI 10.1007/978-1-4757-4242-8_7 CrossRefGoogle ScholarPubMed
Allen, LH. B vitamins in breast milk: relative importance of maternal status and intake, and effects on infant status and function. Adv Nutr. 2012; 3(3), 362369. DOI 10.3945/an.111.001172.CrossRefGoogle ScholarPubMed
Gates, A, Marin, T, De, Leo G, Stansfield, BK. Review of preterm human-milk nutrient composition. Nutr Clin Pract. 2020; 36, 11631172. DOI 10.1002/ncp.10570.CrossRefGoogle ScholarPubMed
Keikha, M, Bahreynian, M, Saleki, M, Kelishadi, R. Macro- and micronutrients of human milk composition: are they related to maternal diet? A comprehensive systematic review. Breastfeed Med. 2017; 12(9), 517527. DOI 10.1089/bfm.2017.0048.CrossRefGoogle ScholarPubMed
Keikha, M, Shayan-Moghadam, R, Bahreynian, M, Kelishadi, R. Nutritional supplements and mother’s milk composition: a systematic review of interventional studies. Int Breastfeed J. 2021; 16(1), 130. DOI 10.1186/s13006-020-00354-0.CrossRefGoogle ScholarPubMed
Skibiel, AL, Hood, WR. Milk matters: offspring survival in Columbian ground squirrels is affected by nutrient composition of mother’s milk. Front Ecol Evol. 2015; 3, 111. DOI 10.3389/fevo.2015.00111.CrossRefGoogle Scholar
Stannard, HJ, Miller, RD, Old, JM. Marsupial and monotreme milk—a review of its nutrient and immune properties. PeerJ. 2020; 8(5), e9335. DOI 10.7717/peerj.9335.Google ScholarPubMed
Zamanillo, R, Sánchez, J, Serra, F, Palou, A. Breast milk supply of microRNA associated with leptin and adiponectin is affected by maternal overweight/obesity and influences infancy BMI. Nutrients. 2019; 11(11), 2589. DOI 10.3390/nu11112589.Google ScholarPubMed
Munblit, D, Treneva, M, Peroni, DG, et al. Immune components in human milk are associated with early infant immunological health outcomes: a prospective three-country analysis. Nutrients. 2017; 9(6), 532. DOI 10.3390/nu9060532.CrossRefGoogle ScholarPubMed
Alsaweed, M, Hartmann, PE, Geddes, DT, Kakulas, F. Micrornas in breastmilk and the lactating breast: potential immunoprotectors and developmental regulators for the infant and the mother. Int J Environ Res Public Health. 2015; 12(11), 1398114020. DOI 10.3390/ijerph121113981.CrossRefGoogle ScholarPubMed
Le, Doare K, Holder, B, Bassett, A, Pannaraj, PS. Mother’s milk: a purposeful contribution to the development of the infant microbiota and immunity. Front Immunol. 2018; 9, 361. DOI 10.3389/fimmu.2018.00361.Google Scholar
Moossavi, S, Sepehri, S, Robertson, B, et al. Composition and variation of the human milk microbiota are influenced by maternal and early-life factors. Cell Host Microbe. 2019; 25(2), 324335. DOI 10.1016/j.chom.2019.01.011.CrossRefGoogle ScholarPubMed
Golan-Gerstl, R, Elbaum Shiff, Y, Moshayoff, V, Schecter, D, Leshkowitz, D, Reif, S. Characterization and biological function of milk-derived miRNAs. Mol Nutr Food Res. 2017; 61(10), 1700009. DOI 10.1002/mnfr.201700009.CrossRefGoogle ScholarPubMed
de la Torre Gomez, C, Goreham, RV, Bech Serra, JJ, Nann, T, Kussmann, M. Exosomics”-A review of biophysics, biology and biochemistry of exosomes with a focus on human breast milk. Front Genet. 2018; 9, 92. DOI 10.3389/fgene.2018.00092.CrossRefGoogle ScholarPubMed
Zempleni, J, Aguilar-Lozano, A, Sadri, M, et al. Biological activities of extracellular vesicles and their cargos from bovine and human milk in humans and implications for infants. J Nutr. 2017; 147(1), 310. DOI 10.3945/jn.116.238949.CrossRefGoogle ScholarPubMed
McGuire, MK, Seppo, A, Goga, A, et al. Best practices for human milk collection for COVID-19 research. Breastfeed Med. 2021; 16(1), 2938. DOI 10.1089/bfm.2020.0296.CrossRefGoogle ScholarPubMed
Bernstein, RM, Hinde, K. Bioactive factors in milk across lactation: maternal effects and influence on infant growth in rhesus macaques (Macaca mulatta). Am J Primatol. 2016; 78(8), 838850. DOI 10.1002/AJP.22544.Google Scholar
Puppel, K, Gołębiewski, M, Grodkowski, G, et al. Composition and factors affecting quality of bovine colostrum: a review. Animals. 2019; 9(12), 1070. DOI 10.3390/ani9121070.CrossRefGoogle ScholarPubMed
Boss, M, Gardner, H, Hartmann, P. Normal human lactation: closing the gap. F1000Research. 2018; 7, F1000. DOI 10.12688/F1000RESEARCH.14452.1.CrossRefGoogle ScholarPubMed
Christian, P, Smith, ER, Lee, SE, Vargas, AJ, Bremer, AA, Raiten, DJ. The need to study human milk as a biological system. Am J Clin Nutr. 2021; 113(5), 10631072. DOI 10.1093/AJCN/NQAB075.Google ScholarPubMed
Langley-Evans, SC. Developmental programming of health and disease. Proc Nutr Soc. 2006; 65(1), 97105. DOI 10.1079/pns2005478.CrossRefGoogle ScholarPubMed
Alfaradhi, M, Ozanne, S. Developmental programming in response to maternal overnutrition. Front Genet. 2011; 2, 27. DOI 10.3389/FGENE.2011.00027.CrossRefGoogle ScholarPubMed
Gluckman, PD, Hanson, MA, Pinal, C. The developmental origins of adult disease. Matern Child Nutr. 2005; 1, 130141. DOI 10.1111/j.1740-8709.2005.00020.x.CrossRefGoogle ScholarPubMed
Aizer, A, Stroud, L, Buka, S. Maternal stress and child outcomes: evidence from siblings. J Hum Resour. 2016; 51(3), 523555. DOI 10.3386/W18422.CrossRefGoogle ScholarPubMed
Oftedal, OT. The evolution of milk secretion and its ancient origins. Animal. 2012; 6(3), 355368. DOI 10.1017/S1751731111001935.CrossRefGoogle ScholarPubMed
Robertson, RC, Manges, AR, Finlay, BB, Prendergast, AJ. The human microbiome and child growth – first 1000 days and beyond. Trends Microbiol. 2019; 27(2), 131147. DOI 10.1016/j.tim.2018.09.008.Google ScholarPubMed
Forbes, JD, Azad, MB, Vehling, L, et al. Association of exposure to formula in the hospital and subsequent infant feeding practices with gut microbiota and risk of overweight in the first year of life. JAMA Pediatr. 2018; 172(7), e181161. DOI 10.1001/jamapediatrics.2018.1161.CrossRefGoogle ScholarPubMed
Azad, MB, Konya, T, Maughan, H, et al. Gut microbiota of healthy Canadian infants: profiles by mode of delivery and infant diet at 4 months. Can Med Assoc J. 2013; 185(5), 385394. DOI 10.1503/cmaj.121189.CrossRefGoogle ScholarPubMed
Badillo-Suárez, PA, Rodríguez-Cruz, M, Nieves-Morales, X. Impact of metabolic hormones secreted in human breast milk on nutritional programming in childhood obesity. J Mammary Gland Biol Neoplasia. 2017; 22(3), 171191. DOI 10.1007/s10911-017-9382-y.CrossRefGoogle ScholarPubMed
Mirza, AH, Kaur, S, Nielsen, LB, et al. Breast milk-derived extracellular vesicles enriched in exosomes from mothers with type 1 diabetes contain aberrant levels of micrornas. Front Immunol. 2019; 10, 2543. DOI 10.3389/fimmu.2019.02543.CrossRefGoogle ScholarPubMed
Chen, Y, Wang, J, Yang, S, et al. Effect of high-fat diet on secreted milk transcriptome in midlactation mice. Physiol Genomics. 2017; 49(12), 747762. DOI 10.1152/physiolgenomics.00080.2017.Google ScholarPubMed
Fodor, A, Zelena, D. The effect of maternal stress activation on the offspring during lactation in light of vasopressin. Sci World J. 2014; 2014(5), 115. DOI 10.1155/2014/265394.CrossRefGoogle ScholarPubMed
Monks, J, Orlicky, DJ, Stefanski, AL, et al. Maternal obesity during lactation may protect offspring from high fat diet-induced metabolic dysfunction. Nutr Diabetes. 2018; 8(1), 55. DOI 10.1038/s41387-018-0027-z.CrossRefGoogle ScholarPubMed
Sasaki, A, de Vega, W, St-Cyr, S, Pan, P, McGowan, P. Perinatal high fat diet alters glucocorticoid signaling and anxiety behavior in adulthood. Neuroscience. 2013; 240(Suppl. 1), 112. DOI 10.1016/j.neuroscience.2013.02.044.CrossRefGoogle ScholarPubMed
Winther, G, Elfving, B, Müller, HK, Lund, S, Wegener, G. Maternal high-fat diet programs offspring emotional behavior in adulthood. Neuroscience. 2018; 388(4), 87101. DOI 10.1016/j.neuroscience.2018.07.014.CrossRefGoogle ScholarPubMed
Ellsworth, L, Perng, W, Harman, E, Das, A, Pennathur, S, Gregg, B. Impact of maternal overweight and obesity on milk composition and infant growth. Matern Child Nutr. 2020; 16(3), 12979. DOI 10.1111/mcn.12979.CrossRefGoogle ScholarPubMed
Forno, E, Young, OM, Kumar, R, Simhan, H, Celedón, JC. Maternal obesity in pregnancy, gestational weight gain, and risk of childhood asthma. Pediatrics. 2014; 134(2), e535e546. DOI 10.1542/peds.2014-0439.CrossRefGoogle ScholarPubMed
Chan, D, Goruk, S, Becker, A, et al. Adiponectin, leptin and insulin in breast milk: associations with maternal characteristics and infant body composition in the first year of life. Int J Obes Adv online Publ. 2017; 42(1), 3643. DOI 10.1038/ijo.2017.189.Google ScholarPubMed
Lemas, DJ, Young, BE, Baker, PR, et al. Alterations in human milk leptin and insulin are associated with early changes in the infant intestinal microbiome. Am J Clin Nutr. 2016; 103(5), 12911300. DOI 10.3945/ajcn.115.126375.CrossRefGoogle ScholarPubMed
Davis, EP, Sandman, CA. The timing of prenatal exposure to maternal cortisol and psychosocial stress is associated with human infant cognitive development. Child Dev. 2010; 81(1), 131148. DOI 10.1111/j.1467-8624.2009.01385.x.CrossRefGoogle ScholarPubMed
Browne, PD, Aparicio, M, Alba, C, et al. Human milk microbiome and maternal postnatal psychosocial distress. Front Microbiol. 2019; 10, 2333. DOI 10.3389/fmicb.2019.02333.CrossRefGoogle ScholarPubMed
Pannaraj, PS, Li, F, Cerini, C, et al. Association between breast milk bacterial communities and establishment and development of the infant gut microbiome. JAMA Pediatr. 2017; 171(7), 647654. DOI 10.1001/jamapediatrics.2017.0378.CrossRefGoogle ScholarPubMed
Carlson, AL, Xia, K, Azcarate-Peril, MA, et al. Infant gut microbiome associated with cognitive development. Biol Psychiatry. 2018; 83(2), 148159. DOI 10.1016/J.BIOPSYCH.2017.06.021.CrossRefGoogle ScholarPubMed
Milani, C, Duranti, S, Bottacini, F, et al. The first microbial colonizers of the human gut: composition, activities, and health implications of the infant gut microbiota. Microbiol Mol Biol Rev. 2017; 81(4), 26050. DOI 10.1128/mmbr.00036-17.CrossRefGoogle ScholarPubMed
Turroni, F, Milani, C, Duranti, S, et al. The infant gut microbiome as a microbial organ influencing host well-being. Ital J Pediatr. 2020; 46(1), 113. DOI 10.1186/s13052-020-0781-0.CrossRefGoogle ScholarPubMed
Bautista, C, Montaño, S, Ramirez, V, et al. Changes in milk composition in obese rats consuming a high-fat diet. Br J Nutr. 2016; 115(3), 538546. DOI 10.1017/S0007114515004547.CrossRefGoogle ScholarPubMed
Panagos, PG, Vishwanathan, R, Penfield-Cyr, A, et al. Breastmilk from obese mothers has pro-inflammatory properties and decreased neuroprotective factors. J Perinatol. 2016; 36(4), 284290. DOI 10.1038/jp.2015.199.CrossRefGoogle ScholarPubMed
Bravi, F, Wiens, F, Decarli, A, Dal Pont, A, Agostoni, C, Ferraroni, M. Impact of maternal nutrition on breast-milk composition: a systematic review. Am J Clin Nutr. 2016; 104(3), 646662. DOI 10.3945/ajcn.115.120881.CrossRefGoogle ScholarPubMed
Innis, SM. Impact of maternal diet on human milk composition and neurological development of infants. Am J Clin Nutr. 2014; 99(3), 734S741S. DOI 10.3945/AJCN.113.072595.CrossRefGoogle ScholarPubMed
Samuel, TM, Zhou, Q, Giuffrida, F, Munblit, D, Verhasselt, V, Thakkar, SK. Nutritional and non-nutritional composition of human milk is modulated by maternal, infant, and methodological factors. Front Nutr. 2020; 7, e0197713. DOI 10.3389/FNUT.2020.576133.CrossRefGoogle ScholarPubMed
Kugananthan, S, Gridneva, Z, Lai, CT, et al. Associations between maternal body composition and appetite hormones and macronutrients in human milk. Nutrients. 2017; 9(3), 252. DOI 10.3390/nu9030252.CrossRefGoogle ScholarPubMed
Young, BE, Patinkin, ZW, Pyle, L, et al. Markers of oxidative stress in human milk do not differ by maternal BMI but are related to infant growth trajectories. Matern Child Health J. 2017; 21(6), 13671376. DOI 10.1007/s10995-016-2243-2.CrossRefGoogle Scholar
Oben, JA, Mouralidarane, A, Samuelsson, AM, et al. Maternal obesity during pregnancy and lactation programs the development of offspring non-alcoholic fatty liver disease in mice. J Hepatol. 2010; 52(6), 913920. DOI 10.1016/j.jhep.2009.12.042.CrossRefGoogle ScholarPubMed
Pomar, CA, Castro, H, Picó, C, Serra, F, Palou, A, Sánchez, J. Cafeteria diet consumption during lactation in rats, rather than obesity per se, alters miR-222, miR-200a, and miR-26a levels in milk. Mol Nutr Food Res. 2019; 63(8), 1800928. DOI 10.1002/mnfr.201800928.CrossRefGoogle ScholarPubMed
Thibeau, S, D’Apolito, K, Minnick, AF, et al. Relationships of maternal stress with milk immune components in African American mothers of healthy term infants. Breastfeed Med. 2016; 11(1), 614. DOI 10.1089/bfm.2015.0117.CrossRefGoogle ScholarPubMed
Chen, DC, Nommsen-Rivers, L, Dewey, KG, Lönnerdal, B. Stress during labor and delivery and early lactation performance. Am J Clin Nutr. 1998; 68(2), 335344. DOI 10.1093/ajcn/68.2.335.CrossRefGoogle ScholarPubMed
Denić, M, Sunarić, S, Genčić, M, et al. Maternal age has more pronounced effect on breast milk retinol and β-carotene content than maternal dietary pattern. Nutrition. 2019; 65(suppl 1), 120125. DOI 10.1016/J.NUT.2019.02.019.CrossRefGoogle ScholarPubMed
Butts, C, Hedderley, D, Herath, T, et al. Human milk composition and dietary intakes of breastfeeding women of different ethnicity from the Manawatu-Wanganui region of New Zealand. Nutrients. 2018; 10(9), 1231. DOI 10.3390/NU10091231.CrossRefGoogle ScholarPubMed
Tricco, AC, Lillie, E, Zarin, W, et al. PRISMA extension for scoping reviews (PRISMA-ScR): checklist and explanation. Ann Intern Med. 2018; 169(7), 467473. DOI 10.7326/M18-0850.CrossRefGoogle ScholarPubMed
Edwards, PD, Lavergne, SG, McCaw, LK, et al. Maternal effects in mammals: broadening our understanding of offspring programming. Front Neuroendocrinol. 2021; 62(2), 100924. DOI 10.1016/J.YFRNE.2021.100924.CrossRefGoogle ScholarPubMed
Théry, C, Witwer, KW, Aikawa, E, et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): a position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J Extracell Vesicles. 2018; 7(1). DOI 10.1080/20013078.2018.1535750.CrossRefGoogle ScholarPubMed
Xi, Y, Jiang, X, Li, R, Chen, M, Song, W, Li, X. The levels of human milk microRNAs and their association with maternal weight characteristics. Eur J Clin Nutr. 2016; 70(4), 445449. DOI 10.1038/ejcn.2015.168.CrossRefGoogle ScholarPubMed
Fujimori, M, França, EL, Morais, TC, Fiorin, V, de Abreu, LC, Honório-França, AC. Cytokine and adipokine are biofactors can act in blood and colostrum of obese mothers. BioFactors. 2016; 43(2), 243250. DOI 10.1002/biof.1339.CrossRefGoogle ScholarPubMed
Hernandez, LL, Grayson, BE, Yadav, E, Seeley, RJ, Horseman, ND. High fat diet alters lactation outcomes: possible involvement of inflammatory and serotonergic pathways. PLoS One. 2011; 7(3), e32598. DOI 10.1371/journal.pone.0032598.CrossRefGoogle Scholar
Liu, J, Smith, MG, Dobre, MA, Ferguson, JE. Maternal obesity and breast-feeding practices among white and black women. Obesity. 2010; 18(1), 175182. DOI 10.1038/oby.2009.182.CrossRefGoogle ScholarPubMed
Purcell, RH, Sun, B, Pass, LL, Power, ML, Moran, TH, Tamashiro, KLK. Maternal stress and high-fat diet effect on maternal behavior, milk composition, and pup ingestive behavior. Physiol Behav. 2011; 104(3), 474479. DOI 10.1016/j.physbeh.2011.05.012.CrossRefGoogle ScholarPubMed
Abuaish, S, Spinieli, RL, McGowan, PO. Perinatal high fat diet induces early activation of endocrine stress responsivity and anxiety-like behavior in neonates. Psychoneuroendocrinology. 2018; 98, 1121. DOI 10.1016/j.psyneuen.2018.08.003.CrossRefGoogle ScholarPubMed
Champagne, F, Diorio, J, Sharma, S, Meaney, MJ. Naturally occurring variations in maternal behavior in the rat are associated with differences in estrogen-inducible central oxytocin receptors. Proc Natl Acad Sci. 2001; 98(22), 1273612741. DOI 10.1073/PNAS.221224598.CrossRefGoogle ScholarPubMed
Myers, M, Brunelli, S, Squire, J, Shindeldecker, R, Hofer, M. Maternal behavior of SHR rats and its relationship to offspring blood pressures. Dev Psychobiol. 1989; 22(1), 2953. DOI 10.1002/DEV.420220104.CrossRefGoogle ScholarPubMed
Buonfiglio, DC, Ramos-Lobo, AM, Freitas, VM, et al. Obesity impairs lactation performance in mice by inducing prolactin resistance. Sci Rep. 2016; 6(1), 225. DOI 10.1038/srep22421.CrossRefGoogle ScholarPubMed
Larnkjær, A, Ong, KK, Carlsen, EM, Ejlerskov, KT, Mølgaard, C, Michaelsen, KF. The influence of maternal obesity and breastfeeding on infant appetite- and growth-related hormone concentrations: the SKOT cohort studies. Horm Res Paediatr. 2018; 90(1), 2838. DOI 10.1159/000490114.Google ScholarPubMed
Lagström, H, Rautava, S, Ollila, H, et al. Associations between human milk oligosaccharides and growth in infancy and early childhood. Am J Clin Nutr. 2020; 111(4), 769778. DOI 10.1093/ajcn/nqaa010.CrossRefGoogle ScholarPubMed
Martin, L, Woo, J, Geraghty, S, et al. Adiponectin is present in human milk and is associated with maternal factors. Am J Clin Nutr. 2006; 83(5), 11061111. DOI 10.1093/AJCN/83.5.1106.CrossRefGoogle ScholarPubMed
Wulster-Radcliffe, M, Ajuwon, K, Wang, J, Christian, J, Spurlock, M. Adiponectin differentially regulates cytokines in porcine macrophages. Biochem Biophys Res Commun. 2004; 316(3), 924929. DOI 10.1016/J.BBRC.2004.02.130.CrossRefGoogle ScholarPubMed
Combs, T, Berg, A, Obici, S, Scherer, P, Rossetti, L. Endogenous glucose production is inhibited by the adipose-derived protein Acrp30. J Clin Invest. 2001; 108(12), 18751881. DOI 10.1172/JCI14120.CrossRefGoogle ScholarPubMed
Yu, X, Rong, SS, Sun, X, et al. Associations of breast milk adiponectin, leptin, insulin and ghrelin with maternal characteristics and early infant growth: a longitudinal study. Br J Nutr. 2018; 120(12), 13801387. DOI 10.1017/S0007114518002933.CrossRefGoogle ScholarPubMed
Savino, F, Lupica, M, Benetti, S, Petrucci, E, Liguori, S, Cordero Di Montezemolo, L. Adiponectin in breast milk: relation to serum adiponectin concentration in lactating mothers and their infants. Acta Paediatr. 2012; 101(10), 10581062. DOI 10.1111/J.1651-2227.2012.02744.X.CrossRefGoogle ScholarPubMed
van Rossem, L, Smit, H, Lentjes, E, et al. Does breast milk adiponectin affect BMI and cardio-metabolic markers in childhood? Br J Nutr. 2019; 121(8), 905913. DOI 10.1017/S0007114519000266.CrossRefGoogle ScholarPubMed
Aparicio, M, Browne, PD, Hechler, C, et al. Human milk cortisol and immune factors over the first three postnatal months: relations to maternal psychosocial distress. PLoS One. 2020; 15(5), e0233554. DOI 10.1371/journal.pone.0233554.CrossRefGoogle ScholarPubMed
Sullivan, E, Hinde, K, Mendoza, S, Capitanio, J. Cortisol concentrations in the milk of rhesus monkey mothers are associated with confident temperament in sons, but not daughters. Dev Psychobiol. 2011; 53(1), 96104. DOI 10.1038/jid.2014.371.CrossRefGoogle Scholar
Dettmer, AM, Murphy, AM, Guitarra, D, et al. Cortisol in neonatal mother’s milk predicts later infant social and cognitive functioning in rhesus monkeys. Child Dev. 2018; 89(2), 525538. DOI 10.1111/cdev.12783.CrossRefGoogle ScholarPubMed
Hinde, K, Skibiel, AL, Foster, AB, Del Rosso, L, Mendoza, SP, Capitanio, JP. Cortisol in mother’s milk across lactation reflects maternal life history and predicts infant temperament. Behav Ecol. 2015; 26(1), 269281. DOI 10.1093/beheco/aru186.Google ScholarPubMed
Hahn-Holbrook, J, Le, TB, Chung, A, Davis, EP, Glynn, LM. Cortisol in human milk predicts child BMI. Obesity. 2016; 24(12), 24712474. DOI 10.1002/oby.21682.CrossRefGoogle ScholarPubMed
Grey, KR, Davis, EP, Sandman, CA, Glynn, LM. Human milk cortisol is associated with infant temperament. Psychoneuroendocrinology. 2013; 38(7), 11781185. DOI 10.1016/j.psyneuen.2012.11.002.CrossRefGoogle ScholarPubMed
Nolvi, S, Uusitupa, H-M, Bridgett, DJ, et al. Human milk cortisol concentration predicts experimentally induced infant fear reactivity: moderation by infant sex. Dev Sci. 2018; 21(4), 12625. DOI 10.1111/desc.12625.CrossRefGoogle ScholarPubMed
Groer, M, Davis, M, Steele, K. Associations between human milk SIgA and maternal immune, infectious, endocrine, and stress variables. J Hum Lact. 2004; 20(2), 153158. DOI 10.1177/0890334404264104.CrossRefGoogle ScholarPubMed
Bunney, PE, Zink, AN, Holm, AA, Billington, CJ, Kotz, CM. Orexin activation counteracts decreases in nonexercise activity thermogenesis (NEAT) caused by high-fat diet. Physiol Behav. 2017; 176(12), 139148. DOI 10.1002/ajp.22544.CrossRefGoogle ScholarPubMed
Carney, MC, Tarasiuk, A, Diangelo, SL, et al. Metabolism-related microRNAs in maternal breast milk are influenced by premature delivery. Pediatr Res. 2017; 82(2), 226236. DOI 10.1038/pr.2017.54.CrossRefGoogle ScholarPubMed
Gay, MCL, Koleva, PT, Slupsky, CM, et al. Worldwide variation in human milk metabolome: indicators of breast physiology and maternal lifestyle? Nutrients. 2018; 10(9), 1151. DOI 10.3390/nu10091151.CrossRefGoogle ScholarPubMed
Su, LL, Thamarai Chelvi, SK, Lim, SL, et al. The influence of maternal ethnic group and diet on breast milk fatty acid composition. Ann Acad Med Singapore. 2010; 39(9), 675679.Google ScholarPubMed
Figure 0

Table 1. Search terms and strategy

Figure 1

Fig. 1. PRISMA-ScR flow chart illustrating screening and eligibility criteria for article selection.

Figure 2

Table 2. Overarching research topics and categorization of the articles included in the final review (N = 40)

Figure 3

Table 3. Non-nutritive bioactive components in milk linking maternal factors with offspring outcomes

Supplementary material: File

Eisha et al. supplementary material

Eisha et al. supplementary material

Download Eisha et al. supplementary material(File)
File 34.5 KB