Hostname: page-component-8448b6f56d-sxzjt Total loading time: 0 Render date: 2024-04-19T07:51:20.184Z Has data issue: false hasContentIssue false

Nutrition, infection and stunting: the roles of deficiencies of individual nutrients and foods, and of inflammation, as determinants of reduced linear growth of children

Published online by Cambridge University Press:  23 January 2017

D. Joe Millward*
Affiliation:
Department of Nutritional Sciences, School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, UK
*
*Corresponding author: D. Joe Millward, email D.Millward@surrey.ac.uk
Rights & Permissions [Opens in a new window]

Abstract

The regulation of linear growth by nutritional and inflammatory influences is examined in terms of growth-plate endochondral ossification, in order to better understand stunted growth in children. Linear growth is controlled by complex genetic, physiological, and nutrient-sensitive endocrine/paracrine/autocrine mediated molecular signalling mechanisms, possibly including sleep adequacy through its influence on growth hormone secretion. Inflammation, which accompanies most infections and environmental enteric dysfunction, inhibits endochondral ossification through the action of mediators including proinflammatory cytokines, the activin A-follistatin system, glucocorticoids and fibroblast growth factor 21 (FGF21). In animal models linear growth is particularly sensitive to dietary protein as well as Zn intake, which act through insulin, insulin-like growth factor-1 (IGF-1) and its binding proteins, triiodothyronine, amino acids and Zn2+ to stimulate growth-plate protein and proteoglycan synthesis and cell cycle progression, actions which are blocked by corticosteroids and inflammatory cytokines. Observational human studies indicate stunting to be associated with nutritionally poor, mainly plant-based diets. Intervention studies provide some support for deficiencies of energy, protein, Zn and iodine and for multiple micronutrient deficiencies, at least during pregnancy. Of the animal-source foods, only milk has been specifically and repeatedly shown to exert an important influence on linear growth in both undernourished and well-nourished children. However, inflammation, caused by infections, environmental enteric dysfunction, which may be widespread in the absence of clean water, adequate sanitation and hygiene (WASH), and endogenous inflammation associated with excess adiposity, in each case contributes to stunting, and may explain why nutritional interventions are often unsuccessful. Current interventions to reduce stunting are targeting WASH as well as nutrition.

Type
Review Article
Copyright
Copyright © The Authors 2017 

Introduction

The stature of human adults reflects individual genotype and those environmental factors which influence child linear growth and limit the phenotypic expression of the genotype. Tanner identified growth as a ‘mirror of the conditions of society’, especially the ‘nutritional and hygienic status’ of the population( Reference Tanner 1 ). In Victorian Britain, the industrial revolution and urbanisation of the population had detrimental effects on child growth, reducing adult height to the extent that a 1904 government report( 2 ) identified ‘urban poverty leading to insufficient food and malnutrition’ as a main cause of ‘ill health, poor physical and mental performance and a general deterioration of the race’. In fact, this report is credited with the subsequent introduction of free school meals and other benefits for poor children and their families. The result was an increase in adult height in British men born between 1900 and 1946 of about 1·25 cm/decade, a secular trend in height which continued in those born between 1946 and 1960 at 0·6 cm/decade( Reference Tanner 1 ). The most recent study of adult height changes in the century between the 1896 and 1996 birth cohorts indicate increases ranging from 20·2 cm in South Korean women to little or no change in adult height in some sub-Saharan African countries and in South Asia, and a current gap of 22–23 cm in men and 20 cm in women between the tallest and shortest adult populations( 3 ). This is an indication that throughout the developing world today the ‘nutritional and hygienic status’ is preventing normal growth.

Poor growth in children is currently defined as inadequate height, weight and weight for height, in relation to growth standards, currently those defined by the WHO( 4 ). Stunting, underweight and wasting describe a height-for-age (HA), weight-for-age (WA) and weight-for-height (WH) ≥2 sd below the median of the relevant standard with severe stunting or wasting at ≥3 sd below the standards. In practice, HA Z score or WH Z score are calculated: the differences between the observed values and the growth standards as a fraction or multiple of the sd of the mean values of the standards. Because this sd increases with age, the absolute HA difference (cm) has been suggested to be more appropriate in terms of identifying the time course of stunting and appropriate periods for intervention( Reference Leroy, Ruel and Habicht 5 ). Thus the levelling off of the HA Z score deficit seen after 24 months in multiregional analyses of stunting up to 5 years is not seen if HA difference deficits are examined( Reference Leroy, Ruel and Habicht 5 ). The argument has been made that in reality there are not distinct populations of normal stunted or severely stunted children but rather a gradation of growth faltering. Thus in India the entire length-for-age Z score/HA Z score distribution is shifted to the left (compared with the WHO standards), indicating that all children, and not only those falling below a specific cut-off, are affected by some degree of growth faltering( Reference de Onis and Branca 6 ).

A stunting syndrome

Globally more children are stunted than wasted, with prevalence rates and burdens in 2013 of 25 % or 161 million stunted preschool children compared with 8 % or 51 million wasted( Reference de Onis and Branca 6 Reference de Onis, Blossner and Borgh 8 ). Prevalence rates are particularly high in sub-Saharan Africa and South Asia but some 7 % or 5 million of children in the developed world are also stunted.

Stunting can begin in utero and intra-uterine growth faltering is a greater problem than previously believed; for example, in India stunting rates are about 20 % at birth, and this may account for about half of growth faltering in Indian children under 5 years of age. After birth average HA Z score among infants in deprived populations continues to decline to a greater or lesser extent in all regions until about 24 months of age, and given the well-documented rapid brain growth in the first 2 years, this early period is also critical for long-term neurodevelopment( Reference de Onis and Branca 6 ). Thus, emphasis on the first 1000 d as a crucial period for intervention is based not only on the magnitude of faltering but also on its long-term impact on adult human capital. After 24 months of age on the basis of HA differences, growth continues to falter in poor environments with no indication of a levelling off( Reference Leroy, Ruel and Habicht 5 ), so that global stunting at 5 years comprises 11·2 % in utero, 60·6 % between birth and 2 years and 28 % between 2 and 5 years. Substantial height catch-up can occur between 24 months and mid-childhood and again between mid-childhood and adulthood, even in the absence of any interventions in some populations as a result of an extended pubertal growth phase especially in adolescent girls( Reference Prentice, Ward and Goldberg 9 ), indicating that adolescence represents an additional window of opportunity during which substantial life-cycle and intergenerational effects can be accrued.

The complexity of the interactions between environmental adverse influences which impair linear growth has been recently reviewed in terms of a ‘stunting syndrome’( Reference Prendergast and Humphrey 10 ) and a somewhat simplified version of this is shown in Fig. 1 ( Reference Leroy, Ruel and Habicht 5 , Reference de Onis and Branca 6 , Reference Prendergast and Humphrey 10 Reference Scrimshaw, Taylor and Gordon 15 ).

Fig. 1 The stunting syndrome. Modified and simplified from Prendergast & Humphrey( Reference Prendergast and Humphrey 10 ). The shaded boxes indicate adverse influences while the unshaded boxes indicate outcomes. Stunting is identified as a cyclical process, often starting in utero, connecting maternal nutrition to an intergenerational cycle of growth failure transmitted across generations through the mother( 11 ). High rates are apparent for at least the first 2 years of postnatal life, hence the identification of the critical window of the first 1000 d( Reference de Onis and Branca 6 ) but growth continues to falter in poor environments with no indication of a levelling off( Reference Leroy, Ruel and Habicht 5 ), resulting in school children and adults of short stature. Mothers with short stature and especially teenagers are more likely to have low-birth-weight babies who are subsequently more likely to have growth failure during childhood( Reference Christian, Lee and Angel 12 ). Potential mechanisms explaining intergenerational effects on linear growth are shared genetic characteristics, epigenetic effects, programming of metabolic changes, the mechanics of a reduced space for fetal growth( Reference de Onis and Branca 6 ) and sociocultural factors such as the intergenerational transmission of poverty and deprivation( Reference Martorell and Zongrone 13 ). Multiple environmental influences contribute to impaired growth including poor maternal and child nutrition throughout the cycle, inadequate breast-feeding and inappropriate complementary feeding( 14 ) together with infectious and inflammatory insults( Reference Scrimshaw, Taylor and Gordon 15 ). These interactions are mutually reinforcing through infection exacerbating any malnutrition, because of appetite suppression and reduced food intake, and any malabsorption reducing nutrient intake, while malnutrition reduces immune defence systems, thereby worsening the adverse influence of infections. The multiple pathological changes marked by linear growth retardation in early life are associated with increased morbidity and mortality, reduced physical, neurodevelopmental and economic capacity and an elevated risk of metabolic disease into adulthood( Reference Prendergast and Humphrey 10 ). IQ, intelligence quotient; SGA, small for gestational age; prem, prematurity; HAZ, height-for-age Z score.

As for the details of the nutritional deficiencies and of the infectious and inflammatory insults which inhibit height growth, the pathogenesis underlying linear growth failure is said to be surprisingly poorly understood( Reference Prendergast and Humphrey 10 ). In the present review the intention is to attempt to examine the importance of both nutritional deficiencies, especially energy, protein, Zn and iodine, and infectious-inflammatory insults in the context of what might be described as a first-principles model of linear growth regulation.

Physiology, cellular biology and endocrinology of linear bone-growth regulation

The growth potential of an individual in height and overall shape, mainly a function of bone growth, is genetically determined and each individual will follow a growth curve canalised in terms of both extent and time course if conditions are favourable( Reference Tanner 16 ). In the context of the present review favourable conditions include a diet which can exert an appropriate regulatory anabolic drive on growth( Reference Millward and Rivers 17 , Reference Millward and Rivers 18 ) and provide necessary substrates, in an environment which presents minimal inflammatory challenges. While mechanical loading can influence bone density, length growth is relatively insensitive to physiological dynamic loading, although direct compressive stress on the growth plate inhibits growth( Reference Villemure and Stokes 19 ). One widely quoted model( Reference Karlberg 20 ) involves three additive and partly superimposed phases of postnatal linear-growth from birth to maturity, i.e. infancy, childhood and puberty (the ICP model). When interrupted by malnutrition or infection there is usually some period of catch-up growth in WH( Reference Ashworth 21 Reference Waterlow 23 ) and in HA( Reference Golden 24 ), i.e. a self-correcting response returning the growth pattern to the individual growth channel.

The nature of the genetic programming is quite complex. Genetic factors are often estimated to account for 80 % of the variation in height and genome-wide association studies in adults of recent European origin indicate hundreds of SNP variants clustered in genomic loci and biological pathways that affect human height( Reference Allen, Estrada and Lettre 25 ). Nevertheless, these identified variants explain only about 10 % of the phenotypic variance, with unidentified common variants of similar effect sizes possibly increasing the overall influence to about 20 % of heritable variation. The genetic programming of the time courses of linear-growth, especially in relation to events in the growth plate which mediate the slowing down of the initial very rapid fetal, early infancy growth phase with eventual cessation of linear growth after puberty, is particularly complex( Reference Lui and Baron 26 ), as are the mechanisms which link linear growth to the growth of other organs and tissues. The progressive decline in cellular proliferation throughout the organism may result from a programmed down-regulation of a large set of growth-promoting genes( Reference Lui and Baron 26 ).

However, non-genetic mechanisms also exist to coordinate growth throughout the organism. A protein-stat model of growth control accounts for the accumulation of protein-containing structures, in which dietary protein provides key regulatory and permissive (substrate) roles( Reference Millward 27 ). Within this model, the overall metabolic demand for amino acids for lean tissue growth is linked to dietary intake through an aminostatic appetite mechanism which enables protein intake to meet the demand. Regulation of the demand involves a mechanism in which amino acid intake exerts a largely endocrine-mediated anabolic drive on the growth plate of the long bones which in turn, through passive stretching, activates growth of associated muscles at the level of muscle connective tissue synthesis and myofibrillar protein deposition. This ensures that skeletal muscle growth occurs at a rate and time course which ensures sufficient muscle mass and strength to allow development of body function with increasing bone length and associated height, and the protein deposition in muscle signals increases in appetite. Thus the genetically programmed postnatal height-growth pattern is enabled by sufficient and appropriate dietary protein and other key nutrients, which initiate the endocrine-mediated anabolic drive on the growth plate of the long bones and permit linked muscle growth. This hierarchical scheme is shown in Fig. 2 ( Reference Millward 27 ).

Fig. 2 Physiological coordination of whole-body growth. The hierarchy of height-growth control involves bone length growth, regulating skeletal muscle growth, which influences whole-body energy expenditure, consequent food intake and the size of the visceral organ mass. See Millward( Reference Millward 27 ) for details, including the molecular basis of the muscle growth response to passive stretch by bone.

Endochondral ossification

The target of the anabolic drive for linear growth of bones, which directly influence height (limb bones, ribs and vertebrae), is endochondral ossification within the growth plate( Reference Kronenberg 28 Reference Lui, Nilsson and Baron 32 ). This starts with the differentiation cascade initiated by stem cell clonal expansion as proliferative chondrocytes, followed by hypertrophy, cartilage matrix secretion and apoptosis which releases angiogenic factors that stimulate vascular invasion and migration of osteoblasts and osteoclasts, leading to remodelling of calcified cartilage and formation of trabecular bone. Synchronously, the diameter of the long bone diaphysis increases by osteoblastic deposition of cortical bone beneath the periosteum, and the marrow cavity expands as a consequence of osteoclastic bone resorption at the endosteal surface. This ordered process mediates linear growth until adulthood. Progression of endochondral ossification and linear growth is regulated by the combined influence of those systemic endocrine and local paracrine/autocrine anabolic influences( Reference Lui, Nilsson and Baron 32 ), acting together with small molecules which include specific amino acids such as leucine( Reference Millward 33 ), and Zn2+, which mediate a signalling cascade via a variety of pathways. Linear growth continues until the growth plates fuse during puberty, but bone mineralisation and consolidation of bone mass continues until peak bone mass is achieved during the third to fourth decade. Considerable progress has been made in unravelling the marked complexity of the regulation of the growth plate( Reference Long and Ornitz 29 ). Here the main features are briefly summarised.

Endocrine regulation

The endocrine signalling of the dietary anabolic drive which acts on the growth plate includes insulin and the growth hormone (GH)–insulin-like growth factor (IGF)-1 axis, a mixed endocrine paracrine–autocrine system, the thyroxine/triiodothyronine (T4/T3) axis, androgens, oestrogens, vitamin D, glucocorticoids, and possibly leptin( Reference Nilsson, Marino and De Luca 34 , Reference Wit and Camacho-Hübner 35 ), making for an extremely complex system which is by no means understood and will only be briefly reviewed here. As far as the insulin–GH–IGF-1 axis is concerned, the Karlberg ICP (infancy, childhood and puberty) model of an insulin/IGF-1-driven fetal infancy phase and a GH/IGF-1-driven childhood phase( Reference Karlberg 20 ) may need reassessment, with several authors arguing that GH has an important role in fetal growth( Reference Waters and Kaye 36 , Reference Harvey and Baudet 37 ). It would appear that: (1) GH can be produced within the fetus both from the pituitary and from other fetal tissues( Reference Waters and Kaye 36 , Reference Harvey and Baudet 37 ) and derives to a limited extent from the placenta( Reference Higgins, Russell and Crossey 38 ), the source of most maternal GH during pregnancy( Reference Higgins, Russell and Crossey 38 , Reference Zeck, Widberg and Maylin 39 ); (2) there is a positive correlation between placental GH levels in pregnant women and the birth weight of their offspring( Reference Zeck, Widberg and Maylin 39 Reference McIntyre, Serek and Crane 41 ); (3) a functional GH receptor is widespread in the human fetus at all stages of development( Reference Waters and Kaye 36 ), including on growth plate chondrocytes( Reference Werther, Haynes and Waters 42 , Reference Fernández-Cancio, Audi and Carrascosa 43 ); and (4) GH increases IGF-1 gene expression in human fetal epiphyseal chondrocytes when conditioned by 1,25-dihydroxyvitamin D3 (1,25(OH)2D3)( Reference Fernández-Cancio, Audi and Carrascosa 43 ).

In the postnatal growth plate, insulin, GH and IGF may have overlapping functions. There can be cross-talk between IGF-1 and insulin at the receptor level( Reference Zhang, He and Tsang 44 ) and post-receptor signalling is identical( Reference Siddle 45 , Reference Guntur and Rosen 46 ), involving the Ras/mitogen-activated protein kinase pathway which signals cell proliferation and the PKB/Akt kinase and mammalian target of rapamycin complex 1 (mTORC1) pathway which stimulates cell growth. Also, although the GH receptor is distinctive, some of the GH Janus activated kinase-2 (JAK2)-initiated signalling is similar to insulin/IGF-1 post-receptor signalling( Reference Carter-Su, Schwartz and Argetsinger 47 , Reference Brooks, Wooh and Tunny 48 ). Because the GH receptor is found on chondrocytes of all zones of the growth plate( Reference Parker, Hegde and Buckley 49 ), GH action is more complex than simply activating expression of IGF-1, bone morphogenetic protein and other genes in resting-zone chondrocytes through the JAK/STAT pathway( Reference Darvin, Joung and Yang 50 ). Also IGF-1, like GH, can stimulate proliferation of resting-zone chondrocytes and chondrocyte hypertrophy( Reference Nilsson, Marino and De Luca 34 ). For IGF-1, both its endocrine action through its mainly hepatic-derived circulating form and its paracrine–autocrine action contributes to postnatal growth( Reference Stratikopoulosa, Szabolcsb and Dragatsisc 51 , Reference Elis, Courtland and Wu 52 ). However, the distinctive feature of IGF-1 action is its regulation through the IGF-binding proteins (IGFBP) 1–6( Reference Clemmons 53 ). These have a variety of functions, with IGFBP-1, -2, -4 and -6 mainly inhibitory by preventing receptor binding. Also IGFBP-1, which is increased in plasma of protein-deficient rats( Reference Jousse, Bruhat and Ferrara 54 ), increases IGF-1 clearance from the plasma and subsequent catabolism( Reference Thissen, Davenport and Pucilowska 55 ). IGFBP-3 can protect IGF-1 by maintaining a circulating ternary complex with IGF-1 and the acid-labile subunit (ALS), a circulating hepatic-derived glycoprotein. ALS functions to prolong the half-life of the IGF-1, IGFBP-3 (or IGFBP-5) binary complexes( Reference Boisclair, Rhoads and Ueki 56 ). IGFBP-5 can enhance IGF-1 activity, including induction of chondrocyte proliferation( Reference Kiepe, Ciarmatori and Hoeflich 57 ). Both IGFBP-3 and -5 can be made by chondrocytes under IGF-1 control( Reference Kiepe, Ciarmatori and Hoeflich 57 ). Interestingly, in the context of a role for Zn in the growth plate, IGFBP-3 possesses a metal-binding domain( Reference Forbes, McCarthy and Norton 58 ), and Zn2+ influences binding of IGF to cell surface-associated IGFBP-3 and -5, possibly affecting IGF bioavailability( Reference McCusker 59 ).

As for thyroid hormones, the thyroid-stimulating hormone (TSH) receptor is expressed in growth plate cartilage and cultured chondrocytes, consistent with evidence of its action to inhibit chondrocyte proliferation( Reference Bassett and Williams 31 ). T3 is widely involved( Reference Bassett and Williams 31 ). Its indirect effects involve enhancing the direct effects of IGF-1 on cartilage and influencing the levels of GH-binding protein, IGF-1, IGF-2, IGFBP-3 and IGF bioactivity( Reference Williams, Robson and Shalet 60 ). It also acts through thyroid hormone receptor (TR)α1, one of three T3 nuclear receptors (the other two TRα2 and TRα3 acting as antagonists) to inhibit proliferation and stimulate prehypertrophic and hypertrophic chondrocyte differentiation. In addition, it has a number of non-genomic actions involving various signal transduction pathways( Reference Bassett and Williams 31 ). Thus, overall thyroid hormone is essential for coordinated progression of endochondral ossification, acting to stimulate genes that control chondrocyte maturation and cartilage matrix synthesis, mineralisation and degradation( Reference Bassett and Williams 31 ).

Both androgens and oestrogen contribute to the pubertal growth spurt( Reference Nilsson, Marino and De Luca 34 ). Androgens can stimulate longitudinal bone growth acting directly on growth plate chondrocytes and indirectly by increased local IGF-1 expression( Reference Nilsson, Marino and De Luca 34 ). Androgens also act in boys after aromatisation to oestrogens which signal through the ER-α receptor to mediate the growth spurt directly and, like androgens, through stimulation of the GH–IGF-1 axis. Eventually with skeletal maturation and epiphyseal fusion, the proliferative capacity of the growth plate chondrocytes is exhausted( Reference Nilsson, Marino and De Luca 34 , Reference Wit and Camacho-Hübner 35 ).

Finally, vitamin D is involved in endochondral ossification, although this is poorly understood in terms of its direct as opposed to indirect influence, and its influence may be subtle. Although chondrocytes contain both the vitamin D receptor (at least in hypertrophic but not proliferating chondrocytes( Reference Wang, Zhu and DeLuca 61 )) and can produce 1,25(OH)2D, the major skeletal manifestations of vitamin D deficiency, rickets and osteomalacia, can be corrected by increasing the intestinal absorption of Ca and phosphate, indicating the importance of indirect effects( Reference Bikle 62 ). However, direct effects are observed in the human fetal epiphyseal growth plate with 1,25(OH)2D, like T3, a potent inhibitor of chondrocyte proliferation but enhancing chondrocyte differentiation in terms of stimulating expression of several GH–IGF axis genes such as IGF-1, IGFBP-3 and GHR( Reference Fernández-Cancio, Audi and Carrascosa 43 ). Vitamin D also participates with parathyroid hormone (PTH) in a functional paracrine feedback loop in the growth plate between 1,25(OH)2D and PTH-related protein (PTHrP). Thus 1,25(OH)2D decreases PTHrP production, while PTHrP increases chondrocyte sensitivity to 1,25(OH)2D by increasing vitamin D receptor production( Reference Bach, Rutten and Hendriks 63 ).

The main inhibitory endocrine influence is cortisol, which is increased in energy deficiency and inflammation and its action is discussed below (see the Inflammation and endochondral ossification section).

Paracrine signalling within the growth plate

The linkage between nutritionally mediated endocrine changes in insulin, GH/IGF-1, T3 androgens and oestrogens, as described above, and endochondral ossification is complex and not entirely understood. A brief summary of those key factors involved is reported here( Reference Kronenberg 28 , Reference Long and Ornitz 29 , Reference Bassett and Williams 31 , Reference Xie, Zhou and Chen 64 ). Chondrogenesis is initiated by one of the bone morphogenetic protein (BMP) family of signalling molecules, together with the SOX series of transcription factors. Chondrocyte proliferation is directly stimulated by Indian hedgehog, which ensures that sufficient chondrocyte proliferation occurs by increasing PTHrP signalling, which in turn inhibits chondrocyte hypertrophic differentiation. Wingless/integrated protein signalling promotes hypertrophic differentiation, whereas fibroblast growth factors (FGF) FGF18 and 19, acting through FGFR3, inhibit chondrocyte proliferation and differentiation and stimulate their apoptosis. Epidermal growth factor, vascular endothelial growth factor and transforming growth factor (TGF)-β also play a part, most probably with additive effects as with the additive effects of IGF-1 and TGF-β1 observed on human chondrocytes in culture( Reference Seifarth, Csaki and Shakibaei 65 ).

One of the principal apparent functions of the endocrine system is to allow rapid growth only when the organism is able to consume abundant and appropriate nutrients. Thus any GH-driven linear growth can only occur when GH can induce IGF-1 and the various other paracrine growth factors as a result of the appropriate dietary signalling from key nutrients, such as amino acids and Zn, to increase growth factor levels. This anabolic drive is apparent in the growing rat in which growth and protein synthesis rates in muscle and bone reflect circulating levels of insulin, IGF-1 and free T3, in turn a function of dietary protein intakes( Reference Jepson, Bates and Millward 66 Reference Yahya, Tirapegui and Bates 71 ), and especially leucine which enhances insulin secretion from islet β-cells as well as having direct tissue effects (see Millward( Reference Millward 33 )). However, in children, length growth is very much slower and saltatory (discontinuous) with 90–95 % of infant development growth-free( Reference Lampl, Veldhuis and Johnson 72 ), and the physiology and endocrinology of this saltatory growth is by no means understood. What is known is that GH secretion, which is pulsatile during the day and night and extremely variable on a day-to-day basis, does appear to relate to height growth velocity, in that synchrony between successive changes in height and GH output is associated with increased stature in healthy children( Reference Gill, Tillmann and Veldhuis 73 ). Furthermore, in infants each saltation appears to follow periods of increased sleep( Reference Lampl and Johnson 74 ), and because from early childhood the onset of sleep is a robust stimulus for GH secretion( Reference Van Cauter and Plat 75 ), this may link sleep quality to length growth. In this case low-quality sleep of children in a poor environment could be part of the aetiology of stunting.

Direct anabolic influences of amino acids and zinc

The extent and nature of direct anabolic influences of amino acids or Zn on endochondral ossification are poorly understood. What is known is that chondrocyte development in cell culture is sensitive to amino acid levels( Reference Ishikawa, Chin and Schalk 76 ) and that in a wide variety of cell types, the major pathway through which amino acids influence cell growth and protein synthesis is as obligatory upstream regulators of the mTORC1 signalling pathway, with leucine as the main effector( Reference Millward 33 , Reference Laplante and Sabatini 77 , Reference Kimball and Jefferson 78 ). Given that mTORC1 signalling has been reported to promote osteoblast differentiation from pre-osteoblasts in mouse primary calvarial cells( Reference Chen and Long 79 ), it can be expected that similar signal-transduction pathways operate in chondrocytes. Dietary leucine is not likely to be limited by poor protein quality since it occurs in high concentrations in most dietary proteins including cereals( Reference Millward 33 ) and its signalling role may simply indicate intake of protein in general. In this context it is the case, as discussed below, that plasma leucine levels are particularly low in stunted children in Malawi( Reference Semba, Shardell and Ashour 80 ).

A role for Zn within the growth plate would be expected given that up to 10 % of human genes code for proteins with Zn-binding domains( Reference Cousins, Liuzzi and Lichten 81 ), with >100 Zn2+-dependent enzymes and >2000 Zn2+-dependent known transcription factors( Reference Andreini, Banci and Bertini 82 ). Labile Zn2+ in extra- and intracellular compartments exerts a wide range of influences on cellular functions with an imbalance in Zn2+ homeostasis linked to dysfunction( Reference Cousins, Liuzzi and Lichten 81 , Reference Prasad 83 ), and a role in growth-related signalling is emerging involving those transporters which mediate intracellular Zn trafficking.

Of the two families of Zn transporters which either reduce (ZnT 1–10) or increase (ZIP 1–14) intracellular Zn, transporter-mediated Zn signalling roles appear to involve the ZIP family( Reference Cousins, Aydemir and Lichten 84 Reference Hojyo, Fukada and Shimoda 87 ). Thus an up-regulation of ZIP8 in human osteoarthritis cartilage increases chondrocyte intracellular Zn2+ levels, activating the metal-regulatory transcription factor 1 (MTF1) which increases matrix-degrading enzymes( Reference Kim, Jeon and Shin 85 ). ZIP13 may be important for bone BMP and TGF-β signalling during chondrocyte maturation on the basis of ZIP13 knockout mice studies and a ZIP13 loss of function mutation in human Ehlers–Danlos syndrome which is associated with short stature( Reference Fukada, Civic and Furuichi 86 ). ZIP14 controls G-protein-coupled receptor (GPCR)-mediated signalling and ZIP14 knock-out mice exhibit growth retardation, attributable to disrupted GPCR signalling in the growth plate and elsewhere( Reference Hojyo, Fukada and Shimoda 87 ). As indicated above, the metal-binding domain of IGFBP-3( Reference Forbes, McCarthy and Norton 58 ) allows Zn2+ to influence binding of IGF to cell surface-associated IGFBP-3 and consequent IGF bioavailability( Reference McCusker 59 ). Taken together, these examples show that the availability of Zn2+ could influence the regulation of growth in diverse ways.

Inflammation and endochondral ossification

Inflammation, which occurs as part of many chronic disease conditions( Reference Wong, Dobie and Altowati 88 ), and to a greater or lesser extent in response to infective insults by bacterial pathogens or immunogenic macromolecules either ingested or translocated across a compromised gut mucosa, is associated with increased and sustained production of pro-inflammatory cytokines, chemokines, adhesion molecules, eicosanoids, NO, oxygen radicals, FGF21( Reference Hanada and Yoshimura 89 , Reference Turner, Nedjai and Hurst 90 ) as well as the activin A–follistatin system( Reference Phillips, de Kretser and Hedger 91 , Reference Sozzani and Musso 92 ). This is usually coupled to both GH and insulin resistance and a reduction in circulating IGF-1 and IGFBP-3( Reference Wong, Dobie and Altowati 88 , Reference Sederquist, Fernandez-Vojvodich and Zaman 93 ). Clinically, low-grade inflammation often results in increased circulating hepatic acute-phase reactants, C-reactive protein (CRP) and α-1 acid glycoprotein, elevations in inflammatory markers in stool such as myeloperoxidase, α-1 antitrypsin, and neopterin, and reduced circulating IGF-1 and IGFBP-3 (for example, Prendergast et al. ( Reference Prendergast, Rukobo and Chasekwa 94 ) and DeBoer et al. ( Reference DeBoer, Scharf and Leite 95 )). In rat models these responses associated with insults such as endotoxaemia( Reference Jepson, Pell and Bates 96 , Reference Hasegawa-Ishii, Inaba and Umegaki 97 ), a parasitic infection( Reference Omwega, Bates and Millward 98 ) or elevated levels of glucocorticoid( Reference Odedra and Millward 99 ) induce a profound insulin resistance( Reference Jepson, Pell and Bates 96 ), and markedly inhibit tissue protein synthesis and growth. Children suffering from specific inflammatory diseases such as inflammatory bowel disease, Crohn’s disease, ulcerative colitis, and juvenile idiopathic arthritis, who exhibit physiologically elevated levels of glucocorticoids and proinflammatory cytokines, usually display abnormal growth patterns as well as delayed puberty( Reference Wong, Dobie and Altowati 88 , Reference Sederquist, Fernandez-Vojvodich and Zaman 93 ). Furthermore, linear growth can be increased in these patients by the inhibition of these cytokines( Reference Wong, Dobie and Altowati 88 , Reference Sederquist, Fernandez-Vojvodich and Zaman 93 ).

Key pro-inflammatory cytokines known to inhibit endochondral ossification include TNFα, IL-1 (particularly IL-1β) and IL-6 (in some( Reference Nakajima, Naruto and Miyamae 100 ) but not all studies( Reference Wong, Dobie and Altowati 88 )). Each of these signals via its specific type I cytokine receptor which are structurally divergent from other cytokine receptor types. High concentrations of these cytokines suppress growth in a dose-dependent manner by decreasing chondrocyte proliferation and hypertrophy while increasing apoptosis( Reference Sederquist, Fernandez-Vojvodich and Zaman 93 ). TNFα, a potent mediator of the inflammatory action of the innate immune system, is secreted mainly from activated macrophages, and by many other cell types in response to endotoxins( Reference Wajant, Pfizenmaier and Scheurich 101 ). It induces cytokine production, activation and/or expression of adhesion molecules, with additional functions linked with lipid metabolism, coagulation, insulin resistance and endothelial function, and is one of the most important and pleiotropic cytokines, mediating inflammatory and immune responses. TNFα is produced endogenously throughout the growth plate and can inhibit chondrocyte proliferation, especially in combination with IL-1β( Reference Mårtensson, Chrysis and Savendahl 102 ). IL-1β expression is induced mainly in response to microbial molecules and it signals via both mitogen-activated protein kinases and the transcription factor NF-κB, thereby resulting in further pro-inflammatory cytokine expression. IL-1β induces rapid dedifferentiation of chondrocytes in culture( Reference Seifarth, Csaki and Shakibaei 65 ) and acts in synergy with TNFα to inhibit rat longitudinal bone growth in culture( Reference Mårtensson, Chrysis and Savendahl 102 ). IL-6 is a pleiotropic cytokine with a wide range of biological activities( Reference Kishimoto 103 ) involving haematopoiesis, B-cell maturation and T cell activation, differentiation and regulation, the secretion of acute-phase proteins by the liver, which it does in cooperation with IL-1 while inhibiting hepatic IGF-1 production. Most importantly, IL-6 has direct inhibitory effects on growth-plate chondrocytes( Reference Nakajima, Naruto and Miyamae 100 , Reference Fernandez-Vojvodich, Zaman and Savendahl 104 ).

Activin A is released rapidly into the circulation during inflammation, in advance of TNFα or IL-6( Reference Phillips, de Kretser and Hedger 91 ). Activin A and its antagonist follistatin are members of the TGF-β superfamily, deriving from a wide range of resting and activated immune and other cell types including bone cells, and modulating several aspects of the inflammatory response, including release of pro-inflammatory cytokines, NO production and immune cell activity( Reference Phillips, de Kretser and Hedger 91 ). Increased circulating levels of activin A occur in inflammatory conditions such as inflammatory bowel disease and rheumatoid arthritis and during bacterial septicaemia, hepatitis C infection, and trauma( Reference Sozzani and Musso 92 ). Less is known about the role of this system in the growth plate, but both activin A and follistatin are produced by human osteoblasts, and are involved in controlling the extent of mineralisation and the prevention of over-mineralisation of bone tissue( Reference Eijken, Swagemakers and Koedam 105 , Reference Bowser, Herberg and Arounleut 106 ).

FGF21, one of the endocrine-like FGF which is also expressed by chondrocytes, is increased by inflammatory stimuli( Reference Feingold, Grunfeld and Heuer 107 ) and by undernutrition in mice( Reference Kubicky, Wu and Kharitonenkov 108 ), and in humans in late-stage fasting( Reference Fazeli, Lun and Kim 109 ) and sepsis( Reference Gariani, Drifte and Dunn-Siegrist 110 ), and directly inhibits chondrocyte proliferation and differentiation as well as preventing GH-mediated stimulation of chondrocyte proliferation and differentiation( Reference Wong, Dobie and Altowati 88 , Reference Kubicky, Wu and Kharitonenkov 108 , Reference Wu, Levenson and Kharitonenkov 111 ).

Inflammation is also accompanied by elevated levels of cortisol, which inhibits length growth directly in terms of chondrocyte proliferation, hypertrophy and cartilage matrix production( Reference Wong, Dobie and Altowati 88 , Reference Klein 112 ) and by the blockade of anabolic growth factor signalling( Reference Nilsson, Marino and De Luca 34 ). Children treated with corticosteroids for various reasons exhibit impaired growth( Reference Mushtaq and Ahmed 113 ). In the rat, exogenous corticosterone at physiologically elevated plasma levels inhibits longitudinal bone growth within 24 h with total arrest after 4 d, at which time epiphyseal cartilage width had shrunk, proteoglycan synthesis was markedly suppressed but overall protein synthesis was unchanged( Reference Tirapegui, Yayha and Bates 70 , Reference Yahya, Tirapegui and Bates 71 ). One target appears to be apoptosis of proliferative growth-plate chondrocytes( Reference Chrysis, Zaman and Chagin 114 ), particularly activation of the caspase cascade triggering Bax-mediated mitochondrial apoptosis( Reference Zaman, Chrysis and Huntjens 115 ), as well as suppression of the PKB/Akt kinase IGF-1 signalling pathway( Reference Chrysis, Zaman and Chagin 114 ).

Given that some or all of the proinflammatory cytokines and activin A–follistatin are produced within the growth plate( Reference Sederquist, Fernandez-Vojvodich and Zaman 93 , Reference Eijken, Swagemakers and Koedam 105 , Reference Bowser, Herberg and Arounleut 106 ), the implication is that individually they may be involved in normal bone growth regulation with their inhibitory effect resulting from their combined effects and/or at high concentrations. The key question which remains is the profile and thresholds in the changes from normal to increased levels of these various mediators induced by low-grade inflammation, which does inhibit linear growth.

Nutrition and linear growth

Nutrients can be classified according to their influence on linear-growth( Reference Golden 116 , Reference Golden 117 ). Type 1 nutrients, the majority group, have specific functions with easily identifiable deficiency symptoms, of which growth failure may or may not be included. Type 2, the smaller group, have ubiquitous functions and it is more difficult to detect deficiency symptoms apart from growth failure.

Type 1 nutrients

Of the type 1 nutrients, iodine deficiency does influence linear growth, as discussed below. For Fe and vitamin A, intervention studies suggest that linear growth is not affected by their individual deficiency( Reference Ramakrishnan, Nguyen and Martorell 118 ). For the bone minerals, it is usually assumed that low Ca intakes are not responsible for stunting. In animal models, while Ca deficiency results in low bone mineralisation and reduced bone strength, it does not result in reduced bone length( Reference Chen, Hayakawa and Emura 119 ). It has been suggested that slow growth rates of children in many developing countries may represent an adaptation to limited Ca supply and poor bioavailability even after adaptation of losses to match low intakes. However, where these low intakes occur, case reports indicate biochemical signs of hyperparathyroidism, low bone mineral contents, and rickets even in the presence of adequate vitamin D status( Reference Prentice and Bates 120 ). Ca supplementation studies in developing countries mostly found no significant effects on linear growth retardation and studies of bone mineral acquisition in younger children reported no height effects( Reference Prentice, Ginty and Stear 121 ). Interestingly, Ca supplementation for 13 months in adolescents which did increase bone mineral content, increased height in boys but not girls( Reference Stear, Prentice and Jones 122 ), possibly through an interaction between the sex hormone systems and GH/IGF( Reference Prentice, Ginty and Stear 121 ).

Type 2 nutrients

Type 2 nutrients, of which linear growth inhibition is an immediate response to their deficiency, include protein (specifically N and indispensable amino acids), Zn, P and the main electrolytes K, Na and Mg. Of these, there is evidence that deficiencies of protein and Zn can occur in the human diet, especially for populations consuming diets based on starchy roots with little or no animal-source foods (ASF); their potential role in the nutritional aetiology of stunting is discussed below. There is some limited evidence for phosphate deficiency occurring through a dietary lack( Reference Waterlow 23 ), although phosphate deficiency is likely to be very rare. Indeed, average intakes of P and Mg from most diets are usually substantially greater than their biological requirements and, if not, increased absorption and conservation of their endogenous losses may prevent an inadequate dietary supply contributing to the poor linear growth of Third World children( Reference Prentice and Bates 120 ).

Multiple nutrient deficiencies and growth

Limitations in the type 1–type 2 classification need to be recognised. First, some type 1 nutrients can influence growth in situations where the symptoms of their deficiency may be missed, one example being mild iodine deficiency( Reference Zimmermann 123 Reference Zimmermann, Jooste and Mabapa 128 ). Second, endochondral ossification is a complex system involving multiple hormonal and small molecule signalling systems responding to multiple dietary factors. This means that multiple type 1 and type 2 nutrient deficiencies may induce growth effects through interactions of their deficiencies which would only be identifiable by multiple nutrient supplementation studies. Finally the type 1/type 2 concept was developed in relation to postnatal growth and is probably less relevant to fetal growth where maternal deficiencies of many nutrients including those involved in cell division such as folate and vitamin B12 can influence fetal development. In this context it is not surprising, as discussed below, that multiple micronutrient supplementations of pregnant women have been shown to be beneficial for birth outcomes including birth weight.

Linear growth regulation as observed in animal models.

Unlike the human diet in which multiple nutrient deficiencies occur, animal studies allow the role of individual nutrient deficiencies to be examined.

Protein and energy deficiency

Because animal growth is usually rapid, it is very sensitive to dietary protein intake and quality, reflecting a metabolic demand for protein almost entirely driven by deposition in lean tissue. The protein efficiency ratio (PER) rat growth assay became the industry-standard method of assessing dietary protein quality( Reference Sarwar, Peace and Botting 129 ), until it was recognised that the assay markedly overestimated the importance of protein quality in the human diet( Reference Millward 130 ). This means that while rat growth studies enable the mechanisms of the dietary activation of growth to be evaluated, they are much less likely to indicate how variation in dietary protein quality influences linear growth in children with a quite different metabolic demand( Reference Millward 131 ).

The concept of the anabolic drive( Reference Millward and Rivers 18 ) and the protein-stat model of dietary protein-mediated growth control( Reference Millward 27 ) was developed from a detailed study of the nutritional and endocrinological regulation of muscle and bone length growth in the rat( Reference Jepson, Bates and Millward 66 , Reference Yahya, Bates and Millward 67 , Reference Yahya and Millward 69 Reference Yahya, Tirapegui and Bates 71 ). In response to protein deficiency, slowed tibial length growth was apparent by 3 d( Reference Yahya and Millward 69 ), and this was graded according to the extent of dietary protein deficiency, ceasing with a near-protein-free diet. Tibial length growth was restored in a graded way with increasing dietary protein concentration over a wide range of protein intakes.

As for energy deficiency, while food restriction has long been known to inhibit linear growth( Reference Dickerson and McCance 132 ), the specific effects of energy deficiency, independently from protein deficiency, are more difficult to evaluate. Energy restriction in young rats to 25 % of normal intakes or total starvation induced immediate weight loss but maintained tibial length growth over 4 d, albeit at reduced rates( Reference Tirapegui, Yayha and Bates 70 ), although corticosterone treatment inhibited tibial growth immediately. Progressive food restriction (75, 50 and 25 % ad libitum) of diets containing increasing concentrations of protein, to maintain constant protein intakes, arrested body-weight growth in all groups, with weight loss in the severely restricted rats, but allowed some tibial length growth to continue at all levels of energy restriction with only a slow development of inhibition. Taken together, these experiments strongly support a dietary control mechanism for linear growth in which protein intake is the most powerful macronutrient influence, acting to promote high circulating levels of insulin, IGF-1 and free T3, which together with amino acids act on the growth plate to optimise endochondral ossification at the level of protein and proteoglycan synthesis maintaining both ribosomal capacity (RNA:protein ratio) and activity (protein synthesis:RNA)( Reference Jepson, Bates and Millward 66 , Reference Yahya, Bates and Millward 67 , Reference Yahya and Millward 69 , Reference Yahya, Tirapegui and Bates 71 ). The inhibition of length growth by energy deficiency or corticosteroids was dissociated from and preceded inhibition of protein synthesis and 35S uptake, possibly reflecting an elevation of rates of proteolysis and proteoglycan degradation associated with elevated corticosterone levels.

Zinc deficiency

Notwithstanding the importance of Zn for pre- and postnatal development( Reference Hambidge and Krebs 133 ), evaluating the mechanism of its actions in growth control has proved remarkably difficult. Weight and length growth inhibition is the immediate response to Zn deficiency in the rat and its influence on catch-up growth in infants on a high-energy soya-based formula, which slowed unless additional Zn was added, resulted in the analogy of Zn with an essential amino acid( Reference Golden and Golden 134 ), given the large number of Zn2+-dependent enzymes and transcription factors in tissues. Although some very limited initial muscle growth occurs in the severely Zn-deficient (ZD) rat, a normal Zn concentration is maintained, expanding the muscle Zn pool, by drawing on Zn mobilised from bone( Reference Giugliano and Millward 135 , Reference Rossi, Migliaccio and Corsi 136 ). However, the key problem of animal studies of ZD is to allow for the characteristic inhibition of appetite, i.e. cyclic changes in food intake and associated body weight( Reference Giugliano and Millward 135 , Reference Williams and Mills 137 ). Careful pair-feeding studies indicated that ZD specifically inhibited body weight and muscle growth( Reference Giugliano and Millward 135 ), through both a corticosteroid-induced blockade due to anorexia and reduced food intake, and an impaired insulin response to food intake when appetite returns, with insufficient insulin to fully activate the translational phase of protein synthesis( Reference Giugliano and Millward 138 ). This indicates a Zn deficiency-induced impairment of the anabolic drive at a key initial step of insulin production. However, in terms of linear growth, while 28 d of a very-low-Zn (ZD) diet( Reference Rossi, Migliaccio and Corsi 136 ) induced marked differences between ZD and pair-fed animals in bone Zn and bone histology, the reductions in femur weight and length, in circulating IGF-1, and in thicknesses of both the overall growth plate and of the hypertrophic cartilage, differed little between ZD and pair-fed animals. When food intake and serum IGF-1 levels were maintained in ZD rats given megestrol acetate, a synthetic progestin used clinically to correct anorexia, growth inhibition persisted, a relatively unambiguous demonstration of the inhibition of the anabolic signalling by IGF-1 with Zn deficiency( Reference Browning, MacDonald and Thornton 139 ). Subsequent studies with ZD rats( Reference MacDonald, Wollard-Biddle and Browning 140 ) demonstrated the need for Zn in the IGF-1 mediation of cell division in cultured 3T3 cells, possibly at the level of a specific step in Ca-dependent mitogenic signal transduction through the IGF-1 receptor protein kinase C (PKC) pathway( Reference MacDonald 141 ), PKC being a Zn metalloenzyme. It remains to be seen whether similar Zn effects explain Zn’s role in regulating endochondral ossification, and/or whether low growth-plate Zn2+ levels decrease chondrocyte IGF bioavailability through impaired surface-associated IGFBP-3 function given its metal-binding domain( Reference Forbes, McCarthy and Norton 58 , Reference McCusker 59 ).

Iodine deficiency

Although dietary iodine is an essential nutrient for maintenance of the T4/T3 system, animal models of iodine deficiency are problematic. This is because of a well-documented conservation of iodine at the level of T3. Thus rats fed an iodine-deficient diet for long periods( Reference Riesco, Taurog and Larsen 142 ), or for two generations( Reference Moreno-Reyes, Egrise and Boelaert 143 , Reference Ren, Guo and Zhang 144 ), or pigs fed a goitrogenic diet( Reference Spiegel, Bestetti and Rossi 145 ), maintain plasma T3 and some growth although symptoms of hypothyroidism can be observed( Reference Riesco, Taurog and Larsen 142 ). T3 conservation is thought to involve increased iodine uptake into the thyroid, increased T3 secretion from the thyroid and continued extrathyroidal T4 to T3 conversion( Reference Zimmermann and Boelaert 124 ). Because of this, early rat studies involved thyroidectomy to lower T3 levels and such studies showed the slow growth was associated with a reduction in DNA-dependent RNA polymerase activity( Reference Tata and Widnell 146 ), and a loss of the capacity for tissue protein synthesis in terms of ribosomal RNA( Reference Brown, Bates and Holliday 147 ). T3 administration by osmotic minipump revealed a dose–response of growth, muscle protein synthesis and ribosomal RNA to circulating T3 levels( Reference Brown and Millward 148 ), similar to the relationship between plasma T3 levels and muscle ribosomal RNA observed when growth was manipulated by protein deficiency( Reference Jepson, Bates and Millward 66 ). Since these early animal studies, studies in GM mice have been the main source of information on the complex physiological relationship between centrally regulated thyroid status and the peripheral anabolic actions of T3 on the growth plate outlined above( Reference Bassett and Williams 31 ).

Human studies of nutrition and linear growth

Plant-based diets are consumed by many communities with a high prevalence of stunting in developing societies. These are often limited to staples which for most starchy root crops are nutritionally poor and of low protein quality, and with few vegetables or pulses and often little or no ASF. However, for children raised in sanitary environments by economically independent parents, plant-based diets can be nutritionally adequate. Thus, near-normal linear growth of children is observed in developed countries within vegan communities who adopt the principles of protein complementation and supplementation with limiting nutrients such as vitamins A, D and B12 (for example, O’Connell et al. ( Reference O’Connell, Dibley and Sierra 149 )). With non-supplemented, relatively restricted vegan diets such as the macrobiotic diet, retarded linear growth is observed( Reference Dwyer, Andrew and Berkey 150 ), with faltering after 4 months to a rate which is 3·5 cm/year less than normal( Reference Dagnelie, van Dusseldorp and van Staveren 151 , Reference Van Dusseldorp, Arts and Bergsma 152 ).

Most studies of linear-growth failure in the developing world are more difficult to interpret in terms of an uncomplicated nutritionally poor diet because of an associated impoverished environment. In fact, the overall number of interventions aimed in improving height which have been successful is low. In 2001, of the trials of complementary foods since 1988 in fourteen countries( Reference Allen and Gillespie 153 ), only three improved height( Reference Martorell and Klein 154 Reference Walker, Powell and Grantham-McGregor 156 ). Trials conducted since then, examining both individual nutrients and specific foods, are referred to below.

Energy intake

The introduction of the term ‘protein–calorie malnutrition’ in the 1960s resulted in a debate into the extent of a protein, as opposed to a food or energy gap( Reference Hegsted 157 ), and this prompted intervention studies. Undernourished Indian children, consuming a cereal-based diet with some buffalo milk, judged to provide sufficient protein but inadequate energy were given an energy-rich low-protein supplement (310 kcal (1300 kJ) and 3 g protein/d)( Reference Gopalan, Swaminathan and Kumari 158 ). The intervention increased both height and weight gain over 14 months, enabling growth on the 50th percentile of American children. Furthermore, supplementation protected the linear growth of the children from an outbreak of measles during the study, whereas the measles outbreak depressed height gain and induced some weight loss of the non-supplemented children.

A similar conclusion that energy intake was the limiting factor for linear growth was reached following the INCAP longitudinal intervention study of Guatemalan mothers, infants and severely stunted children in 1969–1977( Reference Habicht, Martorell and Rivera 159 ). The provision of either a good-quality high-protein (protein:energy ratio 28 %) supplement (Atole), or a protein-free low-energy supplement (Fresco) as control, each with some micronutrients, resulted in a greater increase in height with Atole than Fresco at 3 years. However, analysis of variation in the actual intakes of the two supplements consumed by both pregnant women and children showed that energy, not protein, intake predicted both birth weight and height at 3 years of age( Reference Habicht, Martorell and Rivera 159 ). This was consistent with the background diets of this community (mainly maize and beans, with tomatoes, sorghum and cassava)( Reference Martorell, Habicht and Rivera 160 ) which provided sufficient protein but inadequate energy intakes. Allen has argued( Reference Allen 161 ) that their very low energy intake explains why Guatemalan children were more stunted that those from Egypt, Mexico and Kenya studied in the Nutrition Collaborative Research Support Program (CRSP). Overall, the Atole-supplemented children showed only small benefits of supplementation and remained severely stunted even though many of them consumed more energy and much more protein than their requirements( Reference Martorell and Klein 154 , Reference Habicht, Martorell and Rivera 159 ). Given their impoverished environment, it is possible that enteric dysfunction as discussed below was an additional important factor limiting growth.

Krebs et al. ( Reference Krebs, Mazariegos and Chomba 162 ) suggested the possibility of energy insufficiency in their meat intervention study in stunted infants and toddlers in which stunting rates increased during the trial (see below under Animal-source foods and linear growth). They gave 293–439 kJ/d (70–105 kcal/d), a very modest amount compared with their energy requirements of up to 3350 kJ/d (800 kcal/d).

Malcolm’s studies of the stunted Bundi children in a mission school in Papua New Guinea( Reference Malcolm 163 Reference Malcolm 165 ) included the testing of energy deficiency. These children and the adults were very severely stunted consuming a nutrient-poor, low-protein-quality diet of mainly sweet potato and taro. Given the bulk and low energy density of this low-fat diet, an energy insufficiency was possible, especially for children in the mission school, who had a restricted three meals/d schedule. They were more stunted than village children who had an unrestricted access to food throughout the day. There was a small but significant increase in height growth with 60 % more energy and protein when the children ate five rather than three meals per d, but no stimulation of height growth with extra energy as margarine, although there was a marked increase in skinfold thickness. This suggests that energy intake was not the limiting factor for height growth in these children. As discussed below (Animal-source foods and linear growth section), a skimmed milk supplement had the greatest effect on height. Malcolm argued that there were few signs of overt deficiency or morbidity other than stunting, with malaria rates low at 3–7 %. However, diarrhoea and dysentery did occur, accounting for a quarter of the admissions to the mission hospital, and given this evidence of infection, Malcolm’s assertion that ‘It is unlikely that disease is a significant factor in the slow linear-growth rates in Bundi’ has been challenged( Reference Garn 166 ).

Taken together, these studies do indicate that inadequate energy intakes can occur and that in some circumstances the energy deficiency is associated with stunting.

Protein and amino acid intake

Contrary to popular belief, the dietary protein requirement of young children is low in terms of the protein:energy ratio of the requirement: the safe level is about 5 % energy for healthy preschool children( 167 , Reference Millward and Jackson 168 ). This means that with the exception of some very low-protein starchy-root staples like plantain, cassava, taro and sweet potato, most diets and especially cereal-based diets provide more than adequate amounts of protein if sufficient is consumed to meet the energy requirement. The challenge is to define the lower limits of the range of protein:energy ratios of intakes which ensures that the genotype in relation to stature can be expressed.

During exclusive breast feeding, length growth is generally considered to be optimal with protein intakes much lower (about 6–7 % protein:energy) than after weaning and at later stages of childhood. Furthermore, the linear growth of healthy, breast-fed or formula-fed infants in the USA during the first year of life does not differ and is independent of protein intake for both groups( Reference Heinig, Nommsen and Peerson 169 ). No difference in length growth in infants fed either breast milk or experimental formulae was observed with protein intakes either higher than or similar to breast milk( Reference Räiha, Fazzolari-Nesci and Cajozzo 170 ). Whether this means that during infancy linear growth is insensitive to dietary signals or that the threshold for dietary proteins’ influence is below the lowest level of intake observed in healthy breast- or formula-fed infants is not known. Current regulatory advice for infant formula in Europe( 171 ) is for a minimum protein content of 1·8 g/100 kcal (4·184 kJ) (protein:energy ratio=0·072) for cows’ and goats’ milk-based formulae (of which protein quality and digestibility are assumed to match breast milk (which is the case)( Reference Macé, Steenhout and Klassen 172 )), and 2·25 g/100 kcal (4·184 kJ) (protein:energy ratio=0·09) for formula based on isolated soya protein because of a slightly lower quality (lower sulfur amino acid content) and digestibility.

In developed societies the change to a diet based on family foods at weaning involves a dramatic increase in protein intake to levels considerably in excess of requirements, for example, 13 % protein energy in Danish infants at 9 months( Reference Hoppe, Molgaard and Thomsen 173 ) with even higher values maintained in many Nordic countries up to puberty( Reference Hörnell, Lagström and Lande 174 ). The consequences of this increase in protein intakes for linear growth and whether any influence is desirable is a complicated issue. A systematic review of protein intakes and growth of children in the Nordic countries up to puberty( Reference Hörnell, Lagström and Lande 174 ) concluded that a higher protein intake in infancy and early childhood is convincingly associated with increased growth and higher BMI in childhood and possibly related to an undesirable earlier puberty. However, some of the studies quoted in support of this indicated no relationship between protein intake at early childhood and body fatness at 10 years expressed as body fat percentage or BMI( Reference Hoppe, Molgaard and Thomsen 173 ). Also, in healthy Danish preschool children, height was positively associated with protein intake from milk but not from meat( Reference Hoppe, Udam and Lauritzen 175 ), a possible example of the ‘milk effect’ on growth as discussed below. Thus the issue of excess protein intakes in early childhood and obesity in late childhood and adult life requires further clarification.

As far as protein quality is concerned, it is not clear how well linear growth can occur in the best circumstances for cereal-based diets with minimal ASF, given the likely dietary limitations of micronutrients and minerals, as well as the suboptimal protein quality. The high protein:energy ratio of cereals, especially wheat, means that their lysine limitation is to some extent offset by the higher protein supply, although digestibility may limit the protein quality of cereals like millet( Reference Millward and Jackson 168 ). In fact, the largely cereal-based diets reported for the rural, urban, slum and tribal communities in India, which exhibit high prevalence rates of stunting, appear adequate in terms of protein and lysine intakes( Reference Swaminathan, Vaz and Kurpad 176 ). Although countries with the lowest protein and lysine intakes on the basis of FAO food balance sheets have the highest prevalence rates of stunting( Reference Ghosh, Suri and Uauy 177 ), they also have the lowest gross domestic product per capita and highest infant mortality rates, indicating a poor environment. This means that it is unwise to suggest, as these authors did, a causal relationship between stunting and protein adequacy and quality of the adult diets.

The older literature does include some lysine supplementation studies, in one case examining child growth and bone density( Reference Mack, Vose and Kinard 178 ). Lysine supplementation of the diets of sub-adolescent orphanage children for 5 months increased growth and bone density compared with supplemented controls. However, total lysine intakes were >5 times their requirement. Thus the changes were most probably pharmacological effects of lysine which is known to increase Ca absorption( Reference Civitelli, Villareal and Agnusdei 179 ).

There is evidence that improving the protein quality of maize can increase height growth. Zein, the main storage protein of maize, is low in lysine and tryptophan, hence its association with pellagra (niacin deficiency). Maize varieties with good agronomic properties have been developed by selective breeding with much higher levels of lysine and tryptophan. One variety is quality protein maize (QPM) in which the protein content is unchanged but the concentrations of lysine, tryptophan, the sulfur amino acids and threonine are increased by 50, 75, 90 and 40 %, respectively. The adequacy of QPM protein content and quality for child linear growth was demonstrated in rapidly growing infants recovering from malnutrition( Reference Graham, Lembcke and Morales 180 ). When fed as the sole source of dietary protein, fat and energy (but with micronutrient and mineral supplementation), it enabled height growth comparable with that observed with a cows’ milk formula( Reference Graham, Lembcke and Morales 180 ). Although the introduction of QPM has been relatively slow, a recent meta-analysis of community-based studies in maize-eating populations( Reference Nilupa, Gunaratna and De Groote 181 ) showed modest positive effects of QPM on both weight gain (12 % increase) and height gain (9 % increase) compared with conventional maize in infants and young children with mild to moderate undernutrition.

Serum concentrations of most (sixteen out of nineteen) amino acids were low in stunted preschool children from rural Malawi with no evidence of severe acute malnutrition, compared with non-stunted children( Reference Semba, Shardell and Ashour 80 ). Leucine and the other essential amino acids were particularly low( Reference Semba, Shardell and Ashour 80 ). While this suggests that dietary protein intake was low, without any indication of the timing of the blood sampling in relation to their last meal it would be unwise to over-interpret the findings.

Taken together, these studies provide some suggestion that height growth in children is sensitive to dietary protein and specifically dietary indispensable amino acids intakes as in the animal models, but with the exception of studies with milk protein (discussed below) any influences are small and some of the evidence is indirect. On the other hand, sufficient or even excess dietary protein intake may not be able to prevent the stunting observed in poor communities. Thus the Nutrition Collaborative Research Support Programme (CRSP) found that linear growth faltering was prevalent in preschool children in Egypt, Kenya and Mexico, even though their intakes of protein and essential amino acids were adequate( Reference Beaton, Calloway and Murphy 182 ).

Zinc intake

The prevalence of Zn deficiency is widely believed to be widespread in developing countries because of low intakes of Zn-rich animal products, diets high in phytates, which inhibit Zn absorption, and Zn losses due to diarrhoea. However, few nationally representative surveys of Zn status( Reference de Benoist, Darnton-Hill and Davidsson 183 ) or intakes have been conducted in low-income countries, and clear evidence of its role in the aetiology of stunting has been difficult to demonstrate. Recently, Zn deficiency prevalence has been modelled from both country-specific absorbable Zn content of the national food supply (from national food balance sheet data), and estimates of physiological requirements for absorbed Zn( Reference Wessells and Brown 184 ), finding 17·3 % of the world’s population to be at risk of inadequate Zn intake, with estimates ranging from 7·5 % in high-income regions to 30 % in South Asia. Overall, the prevalence of inadequate Zn intake correlated with the prevalence of stunting in children under 5 years of age, explaining almost a quarter of the between-country variation in stunting.

As for Zn supplementation trials, evidence for their efficacy in terms of pregnancy and birth weight is very mixed, with the majority of studies identifying no influence( Reference Allen and Gillespie 153 ), and Zn supplementation during pregnancy is not included in the most recent Lancet series on Maternal and Child Undernutrition( Reference Bhutta, Das and Rizvi 185 ). For children, benefits of Zn supplementation include reductions in the duration and severity of both diarrhoea and dysentery and acute lower respiratory infections( Reference Allen and Gillespie 153 , Reference Yakoob, Theodoratou and Jabeen 186 ), all of which may indirectly influence height growth. As for linear growth, results of randomised controlled trials of Zn supplementation for children have been mixed, as might be expected given the limitations of Zn supplementation trials: i.e. difficulty of discerning baseline status in supplemented populations, low bioavailability of supplements, low adherence and side effects, etc. Thus although a 1997 meta-analysis indicated a small, but highly significant, impact of Zn supplements in stunted children( Reference Brown, Peerson and Allen 187 ), more recent meta-analyses have found no influence( Reference Ramakrishnan, Nguyen and Martorell 118 ), a significant positive effect in children under 5 years of age( Reference Imdad and Bhutta 188 ), and, in the case of a Cochrane review( Reference Mayo-Wilson, Junior and Imdad 189 ), moderate-quality evidence of a very small improvement in height in children aged 6 months to 12 years of age which was unlikely to be clinically important.

The small-quantity lipid-based nutrient supplements (discussed below) contain Zn and these have not been shown to be effective in Malawi( Reference Phuka, Maleta and Thakwalakwa 190 Reference Ashorn, Alho and Ashorn 192 ) or in Ghana except as part of a Nutributter supplement( Reference Adu-Afarwuah, Lartey and Brown 193 ). Overall, it seems that for Zn, the initial indications that this could easily be remedied with Zn supplements( Reference Brown, Peerson and Allen 187 ) have yet to be achieved. Nevertheless, it is probably appropriate that in the most recent Lancet Series on Maternal and Child Undernutrition( Reference Bhutta, Das and Rizvi 185 ), preventative Zn supplementation in children is included as an intervention in the Lives Saved Tool modelling, acting both on diarrhoea incidence and stunting to reduce mortality.

Iodine intake

Iodine deficiency is seldom included in discussions of the nutritional aetiology of stunting. Yet even after the widespread introduction of salt-iodination programmes, 2 billion individuals globally may have an insufficient iodine intake, especially in South Asia and sub-Saharan Africa, and 241 million (30 %) of school-age children globally are estimated to have insufficient iodine intakes( Reference Zimmermann 123 , Reference Andersson, Karumbunathan and Zimmermann 125 ).

The health consequence of severe iodine deficiency for human growth and development is clear in terms of delayed physical development through growth arrest and delayed bone maturation, and impaired mental function( Reference Zimmermann and Boelaert 124 , Reference Rohner, Zimmermann and Jooste 126 ). An insufficient iodine supply can be aggravated by goitrogens (glucosinolates, cyanogenic glucosides and some flavonoids) in many plant foods and unclean drinking water, which interfere with thyroid metabolism( Reference Zimmermann 123 , Reference Rohner, Zimmermann and Jooste 126 ). Deficiencies of Se, Fe and vitamin A exacerbate the effects of iodine deficiency. Given the importance of the T4/T3 axis for growth and development, with hypothyroidism decreasing circulating IGF-1 and IGFBP-3 levels, and thyroid hormone replacement increasing them( Reference Miell, Zini and Quin 194 , Reference Iglesias, Bayon and Mendez 195 ) and the fact that severe iodine deficiency results in severe linear-growth retardation, it might be expected that growth faltering would occur in otherwise asymptomatic iodine deficiency. In fact, the evidence for this is scarce. Adaptive changes in iodine and thyroid hormone metabolism occur with iodine deficiency, initially conserving iodine within thyroid tissue in association with increased TSH, and maintaining T4 and T3 levels (subclinical hypothyroidism), eventually followed by reductions in T4 (overt hypothyroidism). Thus, in both severely iodine-deficient and moderately iodine-deficient children, as indicated by their urinary iodine concentration, median T4 concentrations can be in the low–normal range, with only a minority identifiable as hypothyroxinaemic( Reference Zimmermann, Jooste and Mabapa 128 ). Serum total and free T3 concentrations may not decrease until disease is far advanced, because increased TSH concentrations stimulate T3 release from the thyroid( Reference Rohner, Zimmermann and Jooste 126 ). Thus, the enlargement of the thyroid may precede any fall in T3 and any impairment of linear growth. In fact, most randomised controlled trials of iodine supplementation have involved pregnancy, aimed to prevent irreversible brain damage associated with cretinism( Reference Zimmermann 127 ), and, of these, a recent systematic review identified only two studies which reported on child linear growth, observing no improvements( Reference Zhou, Anderson and Gibson 196 ). However, hypothyroid iodine-deficient Columbian children with goitre, short stature and retarded bone age increased in stature when given T4 ( Reference Hernandez-Cassis, Cure-Cure and Lopez-Jaramillo 197 ). Iodine repletion in school-age children who were severely iodine deficient (7- to 10-year-old Moroccan children), moderately iodine deficient (10- to 12-year-old Albanian children) or mildly iodine deficient (5- to 14-year-old South African children) increased IGF-1 levels in each case, and also increased total T4, IGF-1, IGFBP-3 and both WA and HA Z scores in the Moroccan and Albanian children( Reference Zimmermann, Jooste and Mabapa 128 ). This suggests that in communities of low iodine availability where use of iodised salt is limited, if stunting is prevalent, iodine deficiency should be included as a potential part of any nutritional aetiology.

Multiple micronutrient intakes

Given that multiple deficiencies of both type 1 and type 2 nutrients are likely to be common in the diets of many mothers and stunted children, poor growth may reflect interactions of their deficiencies. The recognition of this has resulted in the introduction of multiple micronutrient supplements.

These programmes have developed from an initial focus on Fe, vitamin A, iodine, folate and Zn( 198 ) to multiple-micronutrient supplementations (MMS) of pregnant women in developing countries( 199 ), recommending a single daily tablet containing the ADA (or higher for folic acid) of fifteen micronutrients (vitamins A, B1, B2, B6, B12, C, D, E, niacin, folic acid, Fe, Zn, Cu, I and Se). It has often been given as a powder. A Cochrane review of MMS during pregnancy indicated that it was efficacious for maternal, fetal and infant health outcomes( Reference Haider and Bhutta 200 ): i.e. a reduced frequency of low birth weight, small for gestational age and stillbirths compared with supplements of only Fe, with or without folic acid. However, for postnatal linear growth, while an early MMS 12-month trial in Mexican infants aged 8–14 months showed benefit for the younger children (<12 months)( Reference Rivera, Gonzalez-Cossio and Flores 201 ), a 2009 meta-analysis of multiple micronutrient supplementations of at least three micronutrients identified only a very small significant change in height( Reference Ramakrishnan, Nguyen and Martorell 118 ). Reviews of trials of micronutrient powders given to children under 2 years of age( Reference De-Regil, Suchdev and Vist 202 ) and in children mainly aged 6 months to 6 years of age( Reference Salam, MacPhail and Das 203 ) concluded that micronutrient powders did not increase linear growth. Since these reviews, as discussed below( Reference Krebs, Mazariegos and Chomba 162 ), a multi-micronutrient-fortified cereal supplement given as a control for a multi-country meat intervention trial in stunted infants and toddlers was unable to prevent the worsening of stunting during the trial, even though both Zn and especially Fe status benefitted from the intervention.

The most recent approach is to provide MMS as part of a small-quantity lipid-based nutrient supplements (LNS), for home fortification during complimentary feeding( Reference Arimond, Zeilani and Jungjohann 204 ). This provides about 20 g/120 kcal (502 kJ) of fat (73 % energy of which essential fatty acids are about 50 %), protein at 9 % energy, and aims to be nutritionally complete in terms of both type 1 and type 2 nutrients. Whilst this International Lipid-Based Nutrient Supplements project is ongoing( 205 ), to date they have achieved only very limited success, with a modest linear growth increase in Ghana, but no growth effects in three trials in Malawi. Thus, LNS appear to promote linear growth in some but not all infant populations. The project team suggests that lack of an intervention effect could reflect asymptomatic infections, environmental enteropathy and an unfavourable composition of intestinal bacterial microbiota, restricting linear growth through a multitude of inflammation-related or other pathways in children with adequate dietary intakes( Reference Ashorn, Alho and Ashorn 192 ). As a result, trials of LNS are now underway combined with interventions aimed at water quality, sanitation and hygiene (WASH)( Reference Arnold, Null and Luby 206 , 207 ) in which the nutrition intervention will use a LNS comprising what is described as a next-generation version of Nutributter( Reference Arnold, Null and Luby 206 ).

Animal-source foods and linear growth

ASF are nutrient dense in terms of many highly bioavailable micronutrients and are often more energy dense than plant-based foods through their fat content; thus, they are a good source of fat-soluble vitamins and essential fatty acids. They are also the only source of vitamin B12. It is the case that for many population groups of children, ASF often comprise only a very small part of their diet, if any at all. As discussed above, while diets free of animal products can support near-normal growth, they need to contain a wide variety of plant food sources and to be supplemented with key micronutrients. However, in poor communities in much of the developing world, diets are much less varied and often contain few ASF or energy-dense foods, with ASF culturally unacceptable in some communities.

Several observational studies, for example The Nutrition Collaborative Research Support Program (N/CRSP) in Egypt, Kenya and Mexico, show associations between intakes of ASF and better growth which persisted even after controlling covariates and confounders such as socio-economic status, morbidity, parental literacy and nutritional status( Reference Neumann, Harris and Rogers 208 ). In these studies the greatest deficits in linear growth were found in those with little or no ASF in their diet. On the basis of these studies, Allen & Dror( Reference Allen and Dror 209 ) have widely promoted the strategy of increasing consumption of ASF, i.e. meat, fish, eggs and milk, for improving the amount and bioavailability of micronutrients available to children in the developing world.

As far as meat per se is concerned, few of the interventions associated with better growth identified by Allen & Gillespie( Reference Allen and Gillespie 153 ) involved meat as such and these generally failed to increase height growth or reduce stunting. In a Kenyan trial that supplied school children daily snacks of a local dish (maize, beans and greens) for 2 years with additions of meat, milk or equivalent energy, while all children increased weight compared with no snacks, there was little effect on height, although meat did improve muscle mass and cognitive function more than milk( Reference Neumann, Bwibo and Murphy 210 , Reference Neumann, Murphy and Gewa 211 ). A more recent multicentre 12-month intervention at 6 months of age compared the effect of lyophilised beef with an equi-energetic multiple micronutrient-fortified cereal. This involved infants with an estimated prevalence of stunting of ≥20 %, in rural communities in the Democratic Republic of Congo and Zambia, in semirural communities in the Western Highlands of Guatemala, and in urban communities in Karachi, Pakistan( Reference Krebs, Mazariegos and Chomba 162 ). The micronutrient content of the rice–soya cereal supplement was based on guidelines for multiple micronutrients in complementary foods. Consequently, apart from vitamin B12, dietary levels of minerals and vitamins were higher in the cereal compared with beef supplement, four times higher for Fe, and Fe status at 18 months was better in the cereal group. The design was surprising given that higher indices of Fe status from the meat was a secondary hypothesis. However, the extent of stunting worsened from a length-for-age Z score of –1·4 at baseline to –1·9 at 18 m with no difference between the two groups. In fact, only maternal height and education emerged as (positive) influences on length growth and the authors commented that this may be a surrogate for better socio-economic status and favourable practices such as hygiene and medical care( Reference Krebs, Mazariegos and Chomba 162 ). Importantly, handwashing and use of boiled water for food preparation were emphasised in the trial.

The majority of ASF interventions have involved milk, starting with studies in the 1920–1930s in the UK and India and in the 1950s in USA( Reference Pollock 212 ). Whilst all the studies leave much to be desired in terms of their design and analysis, they resulted in the identification of milk as an important determinant of height growth in children, and they were instrumental in the subsequent legislation in the UK in 1934, 1940 and in 1945 when free school milk was introduced( Reference Atkins 213 ). Since these early studies the influence of cows’ milk intake on height growth post-weaning has been repeatedly confirmed in a large variety of cross-sectional, observational and intervention studies in healthy children in the UK, Denmark, USA and Japan( Reference Hoppe, Mølgaard and Michaelsen 214 Reference Wiley 219 ) and in stunted children in developing societies.

In healthy Danish preschool children, all with protein intakes well above current protein requirements, with >60 % from ASF, height was positively associated with intakes of total animal protein and milk, but not with intakes of vegetable protein or meat( Reference Hoppe, Udam and Lauritzen 175 ). In New Zealand, long-term avoidance of cows’ milk is associated with small stature in pre-pubertal school children which was argued to reflect the simple chronic avoidance of milk, rather than health problems associated with milk allergy or intolerance( Reference Black, Williams and Jones 220 ). In the USA, analysis of National Health and Nutrition Examination Survey (NHANES) 1999–2002 indicated that adult height was positively associated with milk consumption at the ages of 5–12 years and 13–17 years, after controlling for sex, education and ethnicity( Reference Wiley 219 ). However, given the complexity of height growth regulation, it would be very surprising if milk intake explained all the variation and Wiley( Reference Wiley 219 ) reports on five intervention studies in the UK, Switzerland, New Zealand and the USA which report no significant effect on height in mainly adolescent girls. Nevertheless, it is undeniable that milk, which has the function of supporting rapid postnatal growth, can also influence growth throughout childhood and is a determinant of adult height.

At the outset of these studies milk was viewed simply as a nutrient-rich food providing for growth in stature, with elaborations of this view into a ‘Milk hypothesis’ predicting that a greater consumption of milk during infancy and childhood will result in greater stature in adult life( Reference Bogin 218 ). It is the case that milk consumption and lactose digestion after weaning are exclusively human mammalian traits made possible by lactase persistence (LP), the continued production of the enzyme in the post-weaning period into adulthood. LP post-weaning is not the ancestral condition in humans but is made possible by a relatively recent SNP of the LPH (lactase–phlorizin hydrolase) gene( Reference Romero, Mallick and Liebert 221 ). Whilst the emergence of LP was advantageous amongst early pastoralists with a constant source of fresh milk, consumption in industrialised urban societies was very limited until the pasteurisation of the milk supply to cities in the late 19th and early 20th centuries and the widespread use of refrigeration. This means that the marked increase in milk consumption by urban populations in northern Europe and North America is a 20th-century phenomenon.

It has certainly become clear in recent years that because the specific biological function of milk is to enable rapid post-natal growth, it differs in many ways from all other human foods in having growth-promoting properties not found in meat or other ASF, although these properties are by no means understood. Also the widely assumed beneficial role of milk in the human diet after weaning is being questioned( Reference Hoppe, Mølgaard and Michaelsen 214 ). Melnik et al. ( Reference Melnik, John and Schmitz 222 ) identifies the presence of microRNA in milk exosomes which could possibly act as anabolic signals for the stimulation of cellular growth and proliferation. Also, milk has uniquely high levels of amino acids known to be involved in anabolic signaling, especially tryptophan with concentrations in milk proteins uniquely high, 40 % higher than egg and double that of meat. Trytophan is said to activate the GH–IGF-1–mTORC1 pathway either directly or through gastrointestinal glucose-dependent insulinotropic peptide production. The caseins and lactoglobulins contain high concentrations of leucine, although this is not unique to milk, with higher levels in maize and sorghum. As indicated above, leucine is an upstream activator of the mTORC1 signalling pathway, important for cell replication and growth( Reference Laplante and Sabatini 77 ). These features may explain why Hoppe et al. ( Reference Hoppe, Udam and Lauritzen 175 ) showed, in a cross-sectional study of Danish preschool children, that serum IGF-1 levels and overall height were significantly related to milk but not meat intake. They also showed in an intervention study with a high protein supplement of either milk or beef in 8-year-olds that milk, and not meat, increased concentrations of serum IGF-1 and the serum IGF-1:serum IGFBP-3 ratio significantly( Reference Hoppe, Mølgaard and Juul 223 ) and that fasting insulin levels and consequently insulin resistance (calculated with the homeostasis model assessment) were higher in the milk- but not meat-supplemented children( Reference Hoppe, Mølgaard and Vaag 224 ). This combination of increased growth and insulin resistance raises the question of whether these effects of milk in these otherwise healthy children are of benefit or not( Reference Hoppe, Mølgaard and Michaelsen 214 ). The benefit of tallness is said to include a decreased risk of some diseases (cardiovascular and respiratory diseases) but an increased risk of colorectal, postmenopausal breast and ovarian cancers, and possibly pancreatic, prostate and premenopausal breast cancers( 3 ). Furthermore, it has been known for some time that societies with high rates of milk consumption and with tall adults generally exhibit poor bone health in old age with higher fracture rates compared with less developed countries with much less milk consumption( Reference Hegsted 225 ). Why this should occur is not known but the possibility that the more rapid growth of childhood and earlier puberty results in a bone architecture which becomes more fragile in old age deserves investigation.

As for children in developing countries, data on preschool children from seven countries in Central and South America indicate that milk intake was significantly associated with higher HA Z score in all countries, whereas meat and egg/fish/poultry intakes were only associated with height in one of the countries( Reference Ruel 226 ). A comparison of the heights and weights of children of the Ugandan Karamoja, a cattle-herding, milk-consuming people, with those of the Buganda, farmers growing plantain, sweet potatoes and cassava, showed that the Karamoja children exhibited normal height growth but low weight for height, whilst the Bugandan children were stunted but of normal weight for height( Reference Rutishauser and Whitehead 227 ).

Malcolm’s dietary interventions of children of the Bundi people of the highlands of Papua New Guinea with skimmed milk increased their height( Reference Malcolm 163 Reference Malcolm 165 ). The growth rate of the Bundi children, with their diet of mainly sweet potato and taro providing 3 % dietary protein energy, was slower than that of any other reported population in the world( Reference Malcolm 165 ). At 7 years they were 9·2 and 5·7 cm shorter than Guatemalan boys and girls studied in the INCAP study( Reference Martorell, Schroeder and Rivera 228 ). His 3- to 8-month interventions in preadolescents involved three studies comparing their usual diet with supplemental skimmed milk powder or additional energy as margarine or additional food( Reference Lampl, Johnston and Malcolm 164 ). In all three studies some linear growth was observed with the usual diet which was slightly increased by more of the usual diet. However, the growth rate in length was doubled by the skimmed milk in all three experiments. Although the design of these studies leaves much to be desired compared with best current practice they remain unique in terms of the detailed description of the community in which they were carried out( Reference Malcolm 165 ). Nevertheless, the outcome of these studies is consistent with current knowledge of the role of milk in linear growth regulation. As with all such interventions with milk, although these studies are reported as protein supplementations, they provide increases in minerals, phosphate and other key nutrients. For example, from the published composition of skimmed milk powder, extra Zn in Malcolm’s studies would range from 1·2 to 3 mg/d, not insignificant accounts given that most Zn supplementation studies involve 5 mg/d. Extra iodine would range from 40 to 100 µg/d. The WHO recommends a daily intake of iodine of 120 µg for school children( 229 ) so that if these children were iodine deficient, the milk intervention may have met their iodine requirements. Malcolm makes no mention of iodine deficiency in his monograph on the Bundi people( Reference Malcolm 165 ). However, the Highlands of New Guinea were known to be an area of iodine deficiency from the 1960s and were where a classic iodised oil supplementation study occurred( Reference Pharoah and Connolly 230 ), aimed to prevent endemic cretinism. This means that the growth response observed by Malcolm could have reflected an improvement of their protein, Zn and iodine status in addition to any of the other milk effects described above.

Infection and poor linear growth in children

The combined and interactive effects of infections, environmental factors and malnutrition as possible determinants of stunting in children have long been thought to be of great importance but were first articulated in a systematic way by Scrimshaw et al. ( Reference Scrimshaw, Taylor and Gordon 15 ). Both serious acute infections, particularly those that involve the gastrointestinal tract, and chronic infections impair linear growth( Reference Wong, Dobie and Altowati 88 , Reference Stephensen 231 ). Symptomatic infection is common during the first years of life in low-income countries and repeated episodes of diarrhoea or parasitic infection are associated with increased risk of stunting( Reference Arnold, Null and Luby 206 , Reference Guerrant, Oriá and Moore 232 ). As a result of many intervention trials and observational studies, the idea that infectious diseases are the likely causes of a large share of stunting, together with poor diet, is now the mainstream public health view( Reference Bhutta, Das and Rizvi 185 ). The stunting syndrome as illustrated in Fig. 1 identifies interactions between malnutrition and infection throughout the maternal, infant and child life cycle inhibiting growth( Reference Prendergast and Humphrey 10 ). These interactions are mutually reinforcing through infection exacerbating any malnutrition, because of appetite suppression and reduced food intake, and any malabsorption reducing nutrient intake, while malnutrition reduces immune defence systems, thereby worsening the adverse influence of infections.

The relationship between appetite and inflammation in terms of CRP, TNF-α and IL-6 has been clearly identified in adult dialysis patients( Reference Kalantar-Zadeh, Block and McAllister 233 ) and in cancer patients( Reference McMillan, Wigmore and Fearon 234 ), although such relationships would be difficult to detect in stunted children with low-grade inflammation. However, it is very clear that infection and associated inflammation have direct inhibitory influences on anabolic processes throughout the organism, including the growth plate, which are additional to growth inhibition through malnutrition. Some of the evidence in children is discussed above (see the Inflammation and endochondral ossification section above). HIV-infected children exhibit impaired growth, and reduced height velocity is more strongly associated with disease progression than reduced weight velocity( Reference Benjamin, Miller and Benjamin 235 ). While reduced appetite and food intake account for much of the failure of weight growth in such children( Reference Arpadi 236 ), with weight improved by supplemental enteral and tube feedings( Reference Henderson, Saavedra and Perman 237 ) or treatment with megestrol, a progestational agent used as an appetite stimulant( Reference Clarick, Hanekom and Yogev 238 ), linear growth remains depressed in each case.

Endogenous low-grade inflammation associated with increased adiposity may also contribute to stunting in older children and adolescents. Serum TNF-α, IL-6 and CRP concentrations were positively associated with measurements of overweight in children in Europe( Reference Aeberli, Molinari and Spinas 239 ) and among South-African township adolescents, of whom 18 % were stunted, and, of these, more had a body fat percentage above the normal cut-off points than non-stunted adolescents( Reference Kruger, Pretorius and Schutte 240 ). Of the stunted girls, 50 % had excess body fat, serum TNF-α was higher than in non-stunted girls and cluster analysis showed serum IL-6, waist:hip ratio and TNF-α clustered together. While in this cross-sectional study the association between low-grade inflammation and stunting does not indicate causality, it may indicate an inability to catch up from stunting in earlier childhood. In 6- to 24-month-old Brazilian infants, higher levels of high-sensitivity CRP were associated with higher levels of GH and lower levels of IGF-1 and IGFBP-3 (GH resistance), with latter changes associated with shorter stature( Reference DeBoer, Scharf and Leite 95 ). Low-grade inflammation in the first year of life and perturbation of the GH–IGF axis are associated with stunting in an 18-month longitudinal study of apparently healthy Zimbabwean infants( Reference Prendergast, Rukobo and Chasekwa 94 ). The stunting at 18 months reflected both impaired intra-uterine growth, implicating poor maternal health and low in utero IGF-1 levels, and impaired postnatal growth associated with chronic inflammation, starting very early in infancy, supporting the concepts shown in Fig. 1.

Environmental enteric dysfunction

In the context of the stunting syndrome, infection includes specific diagnosed infections such as parasitic infections, especially malaria and intestinal helminths, and diarrhoea, particularly in conditions of poor sanitation and hygiene. One study suggested that 25 % of stunting was attributed to five or more episodes of diarrhoea( Reference Checkley, Buckley and Gilman 241 ). However, while all agree that diarrhoea is implicated as a cause of poor growth, there has been a debate about its relative importance( Reference Bhutta, Ahmed and Black 242 , Reference Solomons, Mazariegos and Brown 243 ). An analysis of seven longitudinal cohort studies conducted in four low-income countries indicated that the average child’s diarrhoea burden between birth and 2 years only accounted for a clinically modest reduction in height growth( Reference Richard, Black and Gilman 244 ). In Gambian infants in which growth faltering in terms of weight and height growth was reported to become apparent in the 2nd year of life, although diarrhoea did occur, its frequency did not explain the growth faltering( Reference Lunn, Northrop-Clewes and Downes 245 ). Direct measurements of an enteropathy associated with increased intestinal permeability, as indicated by the handling of oral lactulose and mannitol, were a much better predictor of impaired weight and height gain. Increased gut permeability implies a failure of normal gut barrier function, which normally prevents translocation of pathogenic organisms and endotoxins, and which can, in extreme cases, be a route to the development of sepsis in patients in intensive care. Subsequent studies in Gambian infants indicated endotoxin translocation in terms of increased plasma concentrations of total IgG and IgG–endotoxin-core antibody (EndoCAb) and these responses were correlated with increased intestinal permeability. In fact, intestinal permeability, plasma IgG concentration and EndoCAb together accounted for 56 % of the growth inhibition( Reference Campbell, Elia and Lunn 246 ). Mucosal biopsies of these children indicated chronic T cell-mediated enteropathy with crypt hyperplasia, villous stunting and high numbers of intra-epithelial lymphocytes in all Gambian children, regardless of nutritional status( Reference Campbell, Murch and Elia 247 ). However, with worsening nutrition, mucosal cytokine production became biased toward a proinflammatory response, i.e. a dominance of interferon-γ over TGF-β expression and with increased TNF-α-producing cells in the mucosa. These authors concluded that in these Gambian children translocation of immunogenic luminal macromolecules across a compromised gut mucosa had resulted in stimulation of systemic immune/inflammatory processes and subsequent growth impairment. On the basis of this and other evidence, Prendergast & Humphrey( Reference Prendergast and Humphrey 10 ) argued that subclinical infection with enteric pathogens is common, even in the absence of diarrhoea( Reference Kotloff, Nataro and Blackwelder 248 ). Humphrey( Reference Humphrey 249 ) identified this subclinical infection due to enteric pathogens as tropical enteropathy, characterised by villous atrophy, crypt hyperplasia, increased permeability, inflammatory cell infiltrate, and modest malabsorption caused by faecal bacteria ingested in large quantities by young children living in conditions of poor sanitation and hygiene. She suggested it to be the primary causal pathway from poor sanitation and hygiene to undernutrition (and by implication stunting), rather than diarrhoea. Subsequently, Korpe & Petri( Reference Korpe and Petri 250 ) reviewed the distinction between this environmental enteropathy and other non-infectious tropical malabsorption syndromes such as tropical sprue, coeliac sprue and Crohn’s disease, and Keusch et al. ( Reference Keusch, Rosenberg and Denno 251 ) introduced the term environmental enteric dysfunction. Its relevance to child health has been recently reviewed( Reference Crane, Jones and Berkley 252 ). Importantly, in the context of the present review, Prendergast & Humphrey( Reference Prendergast and Humphrey 10 ) wrote: ‘We therefore view stunting as an inflammatory disease arising, in part, from primary gut pathology. Since gut damage also occurs with recurrent (especially persistent) diarrhoea, severe acute malnutrition, HIV infection and micronutrient deficiencies, there are multiple overlapping causes of enteropathy in settings of poverty which may exacerbate the growth failure arising from EED (environmental enteric dysfunction)’. The EED pathway between the poor environment and stunting is illustrated in Fig. 3 ( Reference Humphrey 249 Reference Crane, Jones and Berkley 252 ). One potential link between WASH and the development of the enteropathy is a detrimental change in the intestinal microbiota( Reference Crane, Jones and Berkley 252 ), with evidence from longitudinal studies of twin cohorts of stunted children from Malawi and Bangladesh( Reference Gough, Stephens and Moodie 253 ) in whom the relative abundance of Acidaminococcus sp. was shown to be associated with future linear growth deficits. Because Acidaminococcus sp. can utilise glutamate as their sole source of carbon and energy, with glutamate usually a key source of fuel for the healthy enterocyte, the authors speculate that overgrowth of bacteria that can ferment glutamate may impair barrier function with the consequent deleterious effect on linear child growth. Other recent work points to a specific deleterious role of frequent exposure to mycotoxins( Reference Smith, Prendergast and Turner 254 ). These contaminate a wide range of staple foods, including maize and groundnuts, especially aflatoxin, fumonisin and deoxynivalenol. The authors argue that several billion individuals in predominantly developing countries may be at risk of exposure, with epidemiological evidence from multiple countries suggesting that exposure in pregnant women, infant cord blood, and young children is widespread during early life and associated with impaired infant growth velocity. Although aflatoxins have been most widely studied in relation to liver cancer( Reference Bennett and Klich 255 ), all three mycotoxins can mediate intestinal damage through distinct pathways resulting in increased local and systemic pro-inflammatory cytokines with immunomodulation toward an inflammatory state, potentially interfering with the IGF-1 axis. Some years ago Hendrickse et al. ( Reference Hendrickse, Coulter and Lamplugh 256 ) identified a link between aflatoxicosis and kwashiorkor in Sudanese children. Although there was little evidence of aflatoxicosis in malnourished Jamaican children( Reference Golden 257 ), where aflatoxicosis occurs, a link between the induced inflammation and kwashiorkor is consistent with the concept that kwashiorkor involves the catastrophic impact of an external noxious insult on children with inadequate micronutrient protection unable to counter the consequent oxidative stress( Reference Golden 257 ).

Fig. 3 Stunting as a predominantly inflammatory disease resulting from poor hygiene and environmental enteric dysfunction. The pathway between an unsanitary environment, dietary mycotoxins and other potential pathogens, an abnormal intestinal microbiota and stunting includes intestinal inflammation and pathological changes to the GI mucosa. This results in a failure of barrier function allowing translocation of pathogens and endotoxins resulting in a systemic inflammatory response which inhibits bone growth. In addition, nutrient malabsorption occurs exacerbating any dietary insufficiency, worsening malnutrition and increasing susceptibility to infection through its adverse effect on the immune system. This also contributes to the bone growth inhibition. GH, growth hormone; IGF, insulin-like growth factor; IGFBP, insulin-like growth factor binding protein. Adapted from Humphrey( Reference Humphrey 249 ), Korpe & Petri( Reference Korpe and Petri 250 ), Keusch et al. ( Reference Keusch, Rosenberg and Denno 251 ) and Crane et al. ( Reference Crane, Jones and Berkley 252 ).

Recent direct evidence for the EED concept comes from studies in pre-school rural Bangladeshi children( Reference Lin, Arnold and Afreen 258 ). Objective measures of lower water quality, poor sanitary/handwashing infrastructure, lower gut permeability and lower IgG EndoCAb titres were related to lower HA Z score.

One important consequence of a widespread occurrence of EED is that it would explain the largely disappointing nature of nutritional intervention trials: i.e. the fact that only in a very few studies has the provision of supplements of either specific nutrients, multi-nutrient mixes or foods been shown to restore growth to normal or to the increased rates which might be expected if catch-up were occurring. Whilst a review by Dewey & Adu-Afarwuah( Reference Dewey and Adu-Afarwuah 259 ) of thirty-eight reports of such supplementation trials indicated both weight and height gain, the latter in the range of an increase in 0·0–0·64 length-for-age Z score by 12 to 24 months, Humphrey( Reference Humphrey 249 ) commented that none of children studied in these various interventions achieved normal growth. In fact, the height growth achieved in the most successful of these studies was equivalent to only about a third of the average deficit of Asian and African children. Dewey & Adu-Afarwuah( Reference Dewey and Adu-Afarwuah 259 ) did argue that although changes in mean HA Z scores were small, nevertheless the impact on the lower tail of the distribution – that is, on stunting rates per se – could be considerably larger (for example, a fall from 15·8 to 4·7 % in the intervention group). However, they concluded that complementary feeding interventions, by themselves, cannot change the underlying conditions of poverty and poor sanitation that contribute to poor child growth. A typical recent example of the failure of a nutritional intervention is the randomised controlled trial of a meat supplement undertaken by Krebs et al. ( Reference Krebs, Mazariegos and Chomba 162 ) discussed above. The rural, semi-rural and urban communities involves in that study were all likely to be exposed to poor hygiene and may well have suffered from EED. Also as discussed above, the disappointing outcomes of many of the LNS within the International Lipid-Based Nutrient Supplements programme has also been attributed to asymptomatic infections, EED and/or an unfavourable composition of intestinal bacterial microbiota( Reference Ashorn, Alho and Ashorn 192 ).

On the basis of the existing data, two major randomised studies are now investigating the concepts shown in Fig. 3 in terms of the potential role of EED in childhood stunting. WASH Benefits( Reference Arnold, Null and Luby 206 ) is testing, in cluster-randomised trials, improvements in water quality, sanitation, handwashing and child nutrition, alone and in combination, on length growth over the first 12 and 24 months at multiple sites in Bangladesh and Kenya. The Sanitation, Hygiene, Infant Nutrition Efficacy (SHINE) Project( 207 , Reference Mbuya, Tavengwa and Stoltzfus 260 , Reference Desai, Smith and Mbuya 261 ) is a proof-of-concept cluster-randomised trial underway in Zimbabwe testing the protecting of babies from faecal ingestion with a WASH intervention and optimising nutritional adequacy of the infant diet with an infant and young child feeding intervention on length growth and anaemia. In each trial LNS are used, described as a next-generation version of Nutributter, which are identical except that the SHINE trial includes less Fe (6 mg compared with 9 mg) than in WASH Benefits, to mitigate concerns about the potential effect of supplemental Fe on infections( Reference Desai, Smith and Mbuya 261 ).

The role of nutrition and inflammation on the regulation of endochondrial ossification incorporating what has been discussed in this review is illustrated in Fig. 4 ( Reference Karlberg 20 ).

Fig. 4 Role of nutrition and inflammation in the regulation of endochondral ossification. During the three developmental growth phases of the infancy, childhood and puberty (ICP) model of Karlberg( Reference Karlberg 20 ), the primary endocrine activators of growth are insulin (infancy), growth hormone (childhood) and the sex steroids (puberty) (although current evidence suggests that growth hormone may also be involved in the infancy stage). These act on the paracrine/autocrine system within the growth plate which mediate endochondral ossification. However, a suitable nutritional anabolic drive is necessary for this process to occur involving both type I nutrients such as iodine (to enable adequate thyroid hormone production) and type II nutrients of which amino acids and zinc are particularly important in stimulating the endocrine/paracrine system and in having direct regulatory influences, although this is poorly understood at the molecular level. Milk also appears to have a specific influence not observed with other animal-source foods. Elevated levels of glucocorticoids, inhibitory fibroblast growth factors (for example, FGF21), pro inflammatory cytokines, especially TNFα, IL-1β, IL-6 and other inflammatory mediators associated with infection and inflammation and environmental enteric dysfunction as in Fig. 3, block the nutritional anabolic drive and inhibit endochondral ossification. How these interactions between nutrition and infection occur at the cellular and molecular level is poorly understood. IGF, insulin-like growth factor; IGFBP, insulin-like growth factor binding protein; T3, triiodothyronine; 1,25(OH)2D, 1,25-dihydroxyvitamin D; IHH, Indian hedgehog; PTHrP, parathyroid-hormone-related protein; BMP, bone morphogenetic protein; WNT, wingless/integrated protein; VEGF, vascular endothelial growth factor; EED, environmental enteric dysfunction.

Conclusions

It has been commented( Reference de Onis, Blossner and Borgh 8 ) that even though more than a quarter of the world’s children are stunted, stunting often goes unrecognised in children who live in communities where short stature is so common that it seems normal. Nevertheless, for these communities because of the cycle of adverse consequences identified in the stunting syndrome (Fig. 1), few will be able to experience the same sense of wellbeing experienced by many in developed societies. While appropriate activation of growth-plate endochondrial ossification and consequent linear growth require adequate nutrition, of which the influences of dietary energy, iodine, amino acids and Zn and evidence for their deficiencies in the aetiology of stunting have been discussed here, it remains to be discovered what the minimum nutritional requirements are for acceptable linear growth and especially whether the plant-based diets of the most deprived communities can be sufficiently improved to meet such requirements. Assuming that the latest generation of nutritionally complete supplements prove to be effective, their use will not answer this question. As for the nutritional interventions which have improved linear growth, milk stands out in its growth-promoting actions. The evidence of more rapid linear growth and earlier puberty with high milk intakes during childhood suggested by the studies on Scandinavian children raises important but as yet unanswerable questions about the long-term consequences for health within milk-drinking developed communities.

However, it is clear that the dietary anabolic drive on linear growth is seriously compromised in the presence of inflammation, which appears to be widespread. It is encouraging and informative that there are success stories. Thus de Onis et al. ( Reference de Onis, Blossner and Borgh 8 ) point out the dramatic improvements in Brazil’s Northeast where stunting decreased from 34 % in 1986 to 6 % in 2006 as a result of rising incomes and increased access to schools, clean water, sanitation and basic health care. This would reinforce the view of stunting as an inflammatory disease( Reference Prendergast and Humphrey 10 ), in which case public health programmes focusing on WASH in order to minimise environmental enteric dysfunction are urgently required. It is to be hoped that lessons learned from the two WASH and nutrition trials indicated above( Reference Arnold, Null and Luby 206 , 207 ), which include both education about and physical improvements in WASH, will enable cost-effective strategies to be designed.

Acknowledgements

No financial support was received for the preparation of this paper. There are no conflicts of interest.

References

1. Tanner, JM (1992) Growth as a measure of the nutritional and hygienic status of a population. Horm Res 38, Suppl. 1, 106115.Google Scholar
2. Great Britain Inter-Departmental Committee on Physical Deterioration (1904) Report of the Inter- Departmental Committee on Physical Deterioration. London: HM Stationery Office.Google Scholar
3. NCD Risk Factor Collaboration (NCD-RisC) (2016) A century of trends in adult human height. Elife 26, 5 pii: e13410.Google Scholar
4. World Health Organization Multicentre Growth Reference Study Group (2006) WHO Child Growth Standards: length/height-for-age, weight-for-age, weight-for-length, weight-for-height and body mass index-for-age: methods and development. Geneva: WHO. http://www.who.int/childgrowth/standards/Technical_report.pdf (accessed October 2016).Google Scholar
5. Leroy, JL, Ruel, M, Habicht, JP, et al. (2014) Linear growth deficit continues to accumulate beyond the first 1000 days in low- and middle-income countries: global evidence from 51 national surveys. J Nutr 144, 14601466.Google Scholar
6. de Onis, M & Branca, F (2016) Childhood stunting: a global perspective. Mat Child Nutr 12, Suppl. 1, 1226.Google Scholar
7. United Nations Children’s Fund, World Health Organization & World Bank (2012) Joint UNICEF–WHO–The World Bank Child Malnutrition Database: Estimates for 2012 and Launch of Interactive Data Dashboards. http://www.who.int/nutgrowthdb/jme_2012_summary_note_v2.pdf (accessed October 2016).Google Scholar
8. de Onis, M, Blossner, M & Borgh, E (2012) Prevalence and trends of stunting among pre-school children, 1990–2020. Public Health Nutr 15, 142148.Google Scholar
9. Prentice, AM, Ward, KA, Goldberg, GR, et al. (2013) Critical windows for nutritional interventions against stunting. Am J Clin Nutr 97, 911918.Google Scholar
10. Prendergast, AJ & Humphrey, JH (2014) The stunting syndrome in developing countries. Paediatr Int Child Health 34, 250265.Google Scholar
11. United Nations System Standing Committee on Nutrition (2011) 6th Report on the World Nutrition Situation: Progress in Nutrition. United Nations Standing Committee on Nutrition, Geneva, Switzerland. http://www.fao.org/3/a-as211e.pdf (accessed October 2016).Google Scholar
12. Christian, P, Lee, SE, Angel, MD, et al. (2013) Risk of childhood undernutrition related to small-for-gestational age and preterm birth in low- and middle-income countries. Int J Epidemiol 42, 13401355.CrossRefGoogle ScholarPubMed
13. Martorell, R & Zongrone, A (2012) Intergenerational influences on child growth and undernutrition. Paediatr Perinat Epidemiol 26, 302314.CrossRefGoogle ScholarPubMed
14. United Nations Children’s Fund (2011) Infant and Young Child Feeding: Programming Guide. New York: UNICEF.Google Scholar
15. Scrimshaw, NS, Taylor, CE & Gordon, JE (1968) Interactions of nutrition and infection. Monograph Series No. 57. http://apps.who.int/iris/bitstream/10665/41782/1/WHO_MONO_57_(part1).pdf (accessed October 2016).Google Scholar
16. Tanner, JM (1979) A concise history of growth studies from Buffon to Boas. In Human Growth: a Comprehensive Treatise, vol. 3, Neurobiology and Nutrition, pp. 515593 [F Faulkner and JM Tanner, editors]. New York: Plenum Press.CrossRefGoogle Scholar
17. Millward, DJ & Rivers, J (1988) The nutritional role of indispensible amino acids and the metabolic basis for their requirements. Eur J Clin Nutr 42, 367393.Google Scholar
18. Millward, DJ & Rivers, JPW (1989) The need for indispensable amino acids: the concept of the anabolic drive. Diabet Metab Rev 5, 191212.Google Scholar
19. Villemure, I & Stokes, IAF (2009) Growth plate mechanics and mechanobiology. A survey of present understanding. J Biomech 42, 17931803.CrossRefGoogle ScholarPubMed
20. Karlberg, J (1989) A biologically-oriented mathematical model (ICP) for human growth. Acta Paediatr Scand Suppl 350, 7094.Google Scholar
21. Ashworth, A (1974) Ad lib. feeding during recovery from malnutrition. Br J Nutr 31, 109112.Google Scholar
22. Ashworth, A & Millward, DJ (1986) Catch-up growth in children. Nutr Rev 44, 157163.CrossRefGoogle ScholarPubMed
23. Waterlow, JC (2006) Protein Energy Malnutrition. London: Smith–Gordon & Co Ltd.Google Scholar
24. Golden, MHN (1994) Is complete catch-up possible for stunted malnourished children? Eur J Clin Nutr 48, S58S71.Google Scholar
25. Allen, HL, Estrada, K, Lettre, G, et al. (2010) Hundreds of variants clustered in genomic loci and biological pathways affect human height. Nature 467, 832838.Google Scholar
26. Lui, JC & Baron, J (2011) Mechanisms limiting body growth in mammals. Endocr Rev 32, 422440.Google Scholar
27. Millward, DJ (1995) A protein-stat mechanism for the regulation of growth and maintenance of the lean-body mass. Nutr Res Rev 8, 93120.CrossRefGoogle ScholarPubMed
28. Kronenberg, HM (2003) Developmental regulation of the growth plate. Nature 423, 332336.CrossRefGoogle ScholarPubMed
29. Long, F & Ornitz, DM (2013) Development of the endochondral skeleton. Cold Spring Harb Perspect Biol 5, a008334.Google Scholar
30. Xian, CJ (2014) Recent research on the growth plate: regulation, bone growth defects, and potential treatments. J Mol Endocrinol 53, E1E2.Google Scholar
31. Bassett, JHD & Williams, GR (2016) Role of thyroid hormones in skeletal development and bone maintenance. Endocr Rev 37, 135187.CrossRefGoogle ScholarPubMed
32. Lui, JC, Nilsson, O & Baron, J (2014) Recent insights into the regulation of the growth plate. J Mol Endocrinol 53, T1T9.Google Scholar
33. Millward, DJ (2012) Knowledge gained from studies of leucine consumption in animals and humans. J Nutr 142, 2212S2219S.CrossRefGoogle Scholar
34. Nilsson, O, Marino, R, De Luca, F, et al. (2005) Endocrine regulation of the growth plate. Horm Res 64, 157165.Google Scholar
35. Wit, JM & Camacho-Hübner, C (2011) Endocrine regulation of longitudinal bone growth. In Cartilage and Bone Development and its Disorders vol. 21, pp. 3041 [C Camacho-Hübner, O Nilsson and L Sävendahl, editors]. Basel: Karger.CrossRefGoogle Scholar
36. Waters, MJ & Kaye, PL (2002) The role of growth hormone in fetal development. Growth Horm IGF Res 12, 137146.Google Scholar
37. Harvey, S & Baudet, M-L (2014) Extrapituitary growth hormone and growth? Gen Comp Endocrinol 205, 5561.Google Scholar
38. Higgins, MF, Russell, NE, Crossey, PA, et al. (2012) Maternal and fetal placental growth hormone and IGF axis in type 1 diabetic pregnancy. PLOS ONE 7, e29164.CrossRefGoogle ScholarPubMed
39. Zeck, W, Widberg, C, Maylin, E, et al. (2008) Regulation of placental growth hormone secretion in a human trophoblast model – the effects of hormones and adipokines. Pediatr Res 63, 353357.Google Scholar
40. Chellakooty, M, Vangsgaard, K, Larsen, T, et al. (2004) A longitudinal study of intrauterine growth and the placental growth hormone (GH)–insulin-like growth factor I axis in maternal circulation: association between placental GH and fetal growth. J Clin Endocrinol Metab 89, 384391.CrossRefGoogle ScholarPubMed
41. McIntyre, HD, Serek, R, Crane, DI, et al. (2000) Placental growth hormone (GH), GH-binding protein, and insulin-like growth factor axis in normal, growth-retarded, and diabetic pregnancies: correlations with fetal growth. J Clin Endocrinol Metab 85, 11431150.Google Scholar
42. Werther, GA, Haynes, K & Waters, MJ (1993) GH receptors are expressed on human fetal mesenchymal tissues – identification of mRNA and GH binding protein. J Clin Endocrinol Metab 76, 16381646.Google ScholarPubMed
43. Fernández-Cancio, M, Audi, L, Carrascosa, A, et al. (2009) Vitamin D and growth hormone regulate growth hormone/insulin-like growth factor (GH-IGF) axis gene expression in human fetal epiphyseal chondrocytes. Growth Horm IGF Res 19, 232237.Google Scholar
44. Zhang, F, He, Q, Tsang, WP, et al. (2014) Insulin exerts direct, IGF-1 independent actions in growth plate chondrocytes. Bone Res 2, 14012.Google Scholar
45. Siddle, K (2011) Signalling by insulin and IGF receptors: supporting acts and new players. J Mol Endocrinol 47, R1R10.CrossRefGoogle ScholarPubMed
46. Guntur, AR & Rosen, CJ (2013) IGF-1 regulation of key signaling pathways in bone. Bonekey Rep 2, 437.Google Scholar
47. Carter-Su, C, Schwartz, J & Argetsinger, LS (2016) Growth hormone signaling pathways. Growth Horm IGF Res 28, 1115.CrossRefGoogle ScholarPubMed
48. Brooks, AJ, Wooh, JW, Tunny, KA, et al. (2008) Growth hormone receptor; mechanism of action. Int J Biochem Cell Biol 40, 19841989.Google Scholar
49. Parker, EA, Hegde, A, Buckley, M, et al. (2007) Spatial and temporal regulation of GH-IGF-related gene expression in growth plate cartilage. J Endocrinol 194, 3140.Google Scholar
50. Darvin, P, Joung, YH & Yang, YM (2013) JAK2-STAT5B pathway and osteoblast differentiation. JAKSTAT 2, e24931.Google Scholar
51. Stratikopoulosa, E, Szabolcsb, M, Dragatsisc, I, et al. (2008) The hormonal action of IGF1 in postnatal mouse growth. Proc Natl Acad Sci U S A 105, 1937819383.Google Scholar
52. Elis, S, Courtland, H-W, Wu, Y, et al. (2010) Elevated serum levels of IGF-1 are sufficient to establish normal body size and skeletal properties even in the absence of tissue IGF-1. J Bone Miner Res 25, 12571266.Google Scholar
53. Clemmons, DR (1997) Insulin-like growth factor binding proteins and their role in controlling IGF actions. Cytokine Growth Factor Rev 8, 4562.Google Scholar
54. Jousse, C, Bruhat, A, Ferrara, M, et al. (1998) Physiological concentration of amino acids regulates insulin-like-growth-factor-binding protein 1 expression. Biochem J 334, 147153.CrossRefGoogle ScholarPubMed
55. Thissen, JP, Davenport, ML, Pucilowska, JB, et al. (1992) Increased serum clearance and degradation of 125I-labelled IGF-I in protein-restricted rats. Am J Physiol Endocrinol Metab 262, E406E411.Google Scholar
56. Boisclair, YR, Rhoads, RP, Ueki, I, et al. (2001) The acid-labile subunit (ALS) of the 150 kDa IGF-binding protein complex: an important but forgotten component of the circulating IGF system. J Endocrinol 170, 6370.CrossRefGoogle ScholarPubMed
57. Kiepe, D, Ciarmatori, S, Hoeflich, A, et al. (2005) Insulin-like growth factor (IGF)-I stimulates cell proliferation and induces IGF binding protein (IGFBP)-3 and IGFBP-5 gene expression in cultured growth plate chondrocytes via distinct signaling pathways. Endocrinology 146, 30963104.Google Scholar
58. Forbes, BE, McCarthy, P & Norton, RS (2012) Insulin-like growth factor binding proteins: a structural perspective. Front Endocrinol (Lausanne) 3, 38.Google Scholar
59. McCusker, RH (1998) Controlling insulin-like growth factor activity and the modulation of insulin-like growth factor binding protein and receptor binding. J Dairy Sci 81, 17901800.Google Scholar
60. Williams, GR, Robson, H & Shalet, SM (1998) Thyroid hormone actions on cartilage and bone: interactions with other hormones at the epiphyseal plate and effects on linear growth. J Endocrinol 157, 391403.Google Scholar
61. Wang, Y, Zhu, J & DeLuca, HF (2014) Identification of the vitamin D receptor in osteoblasts and chondrocytes but not osteoclasts in mouse bone. J Bone Min Res 29, 685692.Google Scholar
62. Bikle, DD (2012) Vitamin D and bone. Curr Osteoporos Rep 10, 151159.CrossRefGoogle Scholar
63. Bach, FC, Rutten, KR, Hendriks, K, et al. (2014) The paracrine feedback loop between vitamin D3 (1,25(OH)2D3) and PTHrP in prehypertrophic chondrocytes. J Cell Physiol 229, 19992014.Google Scholar
64. Xie, Y, Zhou, S, Chen, H, et al. (2014) Advances in fibroblast growth factor signalling in growth plate development and disorders. J Mol Endocrinol 53, T11T34.Google Scholar
65. Seifarth, C, Csaki, C & Shakibaei, M (2009) Anabolic actions of IGF-I and TGF-β1 on interleukin-1β-treated human articular chondrocytes: evaluation in two and three dimensional cultures. Histol Histopathol 24, 12451262.Google Scholar
66. Jepson, MM, Bates, PC & Millward, DJ (1988) The role of insulin and thyroid hormones in the regulation of muscle growth and protein turnover in response to dietary protein. Br J Nutr 59, 397415.Google Scholar
67. Yahya, ZAH, Bates, PC & Millward, DJ (1990) Responses to protein deficiency of plasma and tissue insulin-like growth factor-I levels and proteoglycan synthesis rates in rat skeletal muscle and bone. J Endocrinol 127, 497503.CrossRefGoogle ScholarPubMed
68. Divino Filho, JC, Hazel, SJ, Anderstam, B, et al. (1999) Effect of protein intake on plasma and erythrocyte free amino acids and serum IGF-I and IGFBP-1 levels in rats. Am J Physiol 277, E693E701.Google Scholar
69. Yahya, ZAH & Millward, DJ (1994) Dietary protein and the regulation of long bone and muscle growth in the rat. Clin Sci 87, 213224.CrossRefGoogle ScholarPubMed
70. Tirapegui, JO, Yayha, ZAH, Bates, PC, et al. (1994) Dietary energy, glucocorticoids, and the regulation of long bone and muscle growth in the rat. Clin Sci 87, 599606.CrossRefGoogle ScholarPubMed
71. Yahya, ZAH, Tirapegui, JO, Bates, PC, et al. (1994) Influence of dietary protein, energy and corticosteroids on protein turnover, proteoglycan sulphation and growth of long bone and skeletal muscle in the rat. Clin Sci 87, 607618.Google Scholar
72. Lampl, M, Veldhuis, JD & Johnson, ML (1992) Saltation and stasis: a model of human growth. Science 258, 801803.CrossRefGoogle Scholar
73. Gill, MS, Tillmann, V, Veldhuis, JD, et al. (2001) Patterns of GH output and their synchrony with short-term height increments influence stature and growth performance in normal children. J Clin Endocrinol Metab 86, 58605863.CrossRefGoogle ScholarPubMed
74. Lampl, M & Johnson, ML (2011) Infant growth in length follows prolonged sleep and increased naps. Sleep 34, 641650.Google Scholar
75. Van Cauter, E & Plat, L (1996) Physiology of growth hormone secretion during sleep. J Pediatr 128, S32S37.Google Scholar
76. Ishikawa, Y, Chin, JE, Schalk, EM, et al. (1986) Effect of amino acid levels on matrix vesicle formation by epiphyseal growth plate chondrocytes in primary culture. J Cell Physiol 126, 399406.Google Scholar
77. Laplante, M & Sabatini, D (2012) mTOR signaling in growth control and disease. Cell 149, 274293.Google Scholar
78. Kimball, SR & Jefferson, LS (2010) Control of translation initiation through integration of signals generated by hormones, nutrients, and exercise. J Biol Chem 285, 2902729032.Google Scholar
79. Chen, J & Long, F (2015) mTORC1 signaling promotes osteoblast differentiation from preosteoblasts. PLOS ONE 10, e0130627.Google Scholar
80. Semba, RD, Shardell, M, Ashour, FAS, et al. (2016) Child stunting is associated with low circulating essential amino acids. EBioMedicine 6, 246252.Google Scholar
81. Cousins, RJ, Liuzzi, JP & Lichten, LA (2006) Mammalian zinc transport, trafficking, and signals. J Biol Chem 281, 2408524089.CrossRefGoogle ScholarPubMed
82. Andreini, C, Banci, L, Bertini, I, et al. (2006) Counting the zinc-proteins encoded in the human genome. J Proteome Res 5, 196201.CrossRefGoogle ScholarPubMed
83. Prasad, AS (1995) Zinc: an overview. Nutrition 11, Suppl. 1, 9399.Google ScholarPubMed
84. Cousins, RJ, Aydemir, TB & Lichten, LA (2010) Transcription factors, regulatory elements and nutrient–gene communication. Proc Nutr Soc 69, 9194.Google Scholar
85. Kim, J-H, Jeon, J, Shin, M, et al. (2014) Regulation of the catabolic cascade in osteoarthritis by the Zn-ZIP8-MTF1 axis. Cell 156, 730743.Google Scholar
86. Fukada, T, Civic, N, Furuichi, T, et al. (2008) The zinc transporter SLC39A13/ZIP13 is required for connective tissue development; its involvement in BMP/TGF-β signaling pathways. PLoS ONE 3, e3642.Google Scholar
87. Hojyo, S, Fukada, T, Shimoda, S, et al. (2011) The zinc transporter SLC39A14/ZIP14 controls G-protein coupled receptor-mediated signaling required for systemic growth. PLoS ONE 6, e18059.CrossRefGoogle ScholarPubMed
88. Wong, SC, Dobie, R, Altowati, MA, et al. (2016) Growth and the growth hormone-insulin like growth factor 1 axis in children with chronic inflammation: current evidence, gaps in knowledge and future directions. Endocr Rev 37, 62110.Google Scholar
89. Hanada, T & Yoshimura, A (2002) Regulation of cytokine signaling and inflammation. Cytokine Growth Factor Rev 13, 413421.Google Scholar
90. Turner, MD, Nedjai, B, Hurst, T, et al. (2014) Cytokines and chemokines: at the crossroads of cell signalling and inflammatory disease. Biochim Biophys Acta 1843, 25632582.Google Scholar
91. Phillips, DJ, de Kretser, DM & Hedger, MP (2009) Activin and related proteins in inflammation: not just interested bystanders. Cytokine Growth Factor Rev 20, 153164.Google Scholar
92. Sozzani, S & Musso, T (2011) The yin and yang of activin A. Blood 117, 50135015.Google Scholar
93. Sederquist, B, Fernandez-Vojvodich, P, Zaman, F, et al. (2014) Impact of inflammatory cytokines on longitudinal bone growth. J Mol Endocrinol 53, T35T44.Google Scholar
94. Prendergast, AJ, Rukobo, S, Chasekwa, B, et al. (2014) Stunting is characterized by chronic inflammation in Zimbabwean infants. PLOS ONE 9, e86928.Google Scholar
95. DeBoer, MD, Scharf, RJ, Leite, AM, et al. (2017) Systemic inflammation, growth factors, and linear growth in the setting of infection and malnutrition. Nutrition 33, 248253.Google Scholar
96. Jepson, MM, Pell, J, Bates, PC, et al. (1986) The effects of endotoxaemia on protein metabolism in skeletal muscle and liver of fed and fasted rats. Biochem J 235, 329336.Google Scholar
97. Hasegawa-Ishii, S, Inaba, M, Umegaki, H, et al. (2016) Endotoxemia-induced cytokine-mediated responses of hippocampal astrocytes transmitted by cells of the brain–immune interface. Sci Rep 6, 25457.CrossRefGoogle ScholarPubMed
98. Omwega, AM, Bates, PC & Millward, DJ (1988) Vitamin E deficiency in the rat and the response to malarial infection. Proc Nutr Soc 47, 12A.Google Scholar
99. Odedra, BR & Millward, DJ (1982) Effect of corticosterone treatment on muscle protein turnover in adrenalectomised rats and diabetic rats maintained on insulin. Biochem J 204, 663672.Google Scholar
100. Nakajima, S, Naruto, T, Miyamae, T, et al. (2009) Interleukin-6 inhibits early differentiation of ATDC5 chondrogenic progenitor cells. Cytokine 47, 9197.Google Scholar
101. Wajant, H, Pfizenmaier, K & Scheurich, P (2003) Tumor necrosis factor signalling. Cell Death Diff 10, 4565.Google Scholar
102. Mårtensson, K, Chrysis, D & Savendahl, L (2004) Interleukin-1β and TNF-α act in synergy to inhibit longitudinal growth in fetal rat metatarsal bones. J Bone Min Res 19, 18051812.Google Scholar
103. Kishimoto, T (2010) IL-6: from its discovery to clinical applications. Int Immunol 22, 347352.Google Scholar
104. Fernandez-Vojvodich, P, Zaman, F & Savendahl, L (2013) Interleukin-6 acts locally on the growth plate to impair bone growth. Ann Rheum Dis 72, e24.CrossRefGoogle ScholarPubMed
105. Eijken, M, Swagemakers, S, Koedam, M, et al. (2007) The activin A–follistatin system: potent regulator of human extracellular matrix mineralization. FASEB J 21, 29492960.Google Scholar
106. Bowser, M, Herberg, S, Arounleut, P, et al. (2013) Effects of the activin A–myostatin–follistatin system on aging bone and muscle progenitor cells. Exp Gerontol 48, 290297.Google Scholar
107. Feingold, KR, Grunfeld, C, Heuer, JG, et al. (2012) FGF21 is increased by inflammatory stimuli and protects leptin-deficient ob/ob mice from the toxicity of sepsis. Endocrinology 153, 26892700.Google Scholar
108. Kubicky, RA, Wu, S, Kharitonenkov, A, et al. (2012) Role of fibroblast growth factor 21 (FGF21) in undernutrition-related attenuation of growth in mice. Endocrinology 153, 22872295.Google Scholar
109. Fazeli, PK, Lun, M, Kim, SM, et al. (2015) FGF21 and the late adaptive response to starvation in humans. J Clin Invest 125, 46014611.Google Scholar
110. Gariani, K, Drifte, G, Dunn-Siegrist, I, et al. (2013) Increased FGF21 plasma levels in humans with sepsis and SIRS. Endocr Connect 2, 146153.Google Scholar
111. Wu, S, Levenson, A, Kharitonenkov, A, et al. (2012) Fibroblast growth factor 21 (FGF21) inhibits chondrocyte function and growth hormone action directly at the growth plate. J Biol Chem 287, 2606026067.Google Scholar
112. Klein, DL (2015) The effect of glucocorticoids on bone and muscle. Osteoporos Sarcopenia 1, 3945.Google Scholar
113. Mushtaq, T & Ahmed, SF (2002) The impact of corticosteroids on growth and bone health. Arch Dis Child 87, 9396.Google Scholar
114. Chrysis, D, Zaman, F, Chagin, AS, et al. (2005) Dexamethasone induces apoptosis in proliferative chondrocytes through activation of caspases and suppression of the AKT-phosphatidylinositol 3ʹ-kinase signaling pathway. Endocrinology 146, 13911397.Google Scholar
115. Zaman, F, Chrysis, D & Huntjens, K (2014) Dexamethasone differentially regulates Bcl-2 family proteins in human proliferative chondrocytes: role of pro-apoptotic Bid. Toxicol Lett 224, 196200.Google Scholar
116. Golden, MH (1988) The role of individual nutrient deficiencies in growth retardation of children as exemplified by zinc and protein. In Linear Growth Retardation in Less Developed Countries, pp. 143163 [JC Waterlow, editor]. New York: Raven Press.Google Scholar
117. Golden, MH (1991) The nature of nutritional deficiency in relation to growth failure and poverty. Acta Paed Scand 374, 95110.Google Scholar
118. Ramakrishnan, U, Nguyen, P & Martorell, R (2009) Effects of micronutrients on growth of children under 5 y of age:meta-analyses of single and multiple nutrient interventions. Am J Clin Nutr 89, 191203.Google Scholar
119. Chen, H, Hayakawa, D, Emura, S, et al. (2002) Effect of low or high dietary Ca on the morphology of the rat femur. Histol Histopathol 17, 11291135.Google Scholar
120. Prentice, A & Bates, CJ (1994) Adequacy of dietary mineral supply for human bone growth and mineralisation. Eur J Clin Nutr 48, Suppl. 1, S161S177.Google ScholarPubMed
121. Prentice, A, Ginty, F, Stear, SJ, et al. (2005) Calcium supplementation increases stature and bone mineral mass of 16- to 18-year-old boys. J Clin Endocrinol Metab 90, 31533161.CrossRefGoogle ScholarPubMed
122. Stear, SJ, Prentice, A, Jones, SC, et al. (2003) Effect of a calcium and exercise intervention on the bone mineral status of 16 –18-y-old adolescent girls. Am J Clin Nutr 77, 985992.Google Scholar
123. Zimmermann, MB (2009) Iodine Deficiency. Endocr Rev 30, 376408.Google Scholar
124. Zimmermann, MB & Boelaert, K (2015) Iodine deficiency and thyroid disorders. Lancet Diabetes Endocrinol 3, 286289.CrossRefGoogle ScholarPubMed
125. Andersson, M, Karumbunathan, V & Zimmermann, MB (2012) Global iodine status in 2011 and trends over the past decade. J Nutr 142, 744750.Google Scholar
126. Rohner, F, Zimmermann, M, Jooste, P, et al. (2014) Biomarkers of nutrition for development – iodine review. J Nutr 144, 1322S1342S.Google Scholar
127. Zimmermann, MB (2012) The effects of iodine deficiency in pregnancy and infancy. Paediatr Perinat Epidemiol 26, Suppl. 1, 108117.Google Scholar
128. Zimmermann, MB, Jooste, PL, Mabapa, NS, et al. (2007) Treatment of iodine deficiency in school-age children increases insulin-like growth factor (IGF)-I and IGF binding protein-3 concentrations and improves somatic growth. J Clin Endocrinol Metab 92, 437442.Google Scholar
129. Sarwar, G, Peace, RW, Botting, HG, et al. (1989) Relationship between amino acid scores and protein quality indices based on rat growth. Plant Foods Hum Nutr 39, 3344.Google Scholar
130. Millward, DJ (2012) Amino acid scoring patterns for protein quality assessment. Br J Nutr 108, S31S43.Google Scholar
131. Millward, DJ (2012) Identifying recommended dietary allowances for protein and amino acids: a critique of the 2007 WHO/FAO/UNU report. Br J Nutr 108, S3S21.Google Scholar
132. Dickerson, JWT & McCance, RA (1961) Severe undernutrition in growing and adult animals. 8. The dimensions and chemistry of the long bones. Br J Nutr 15, 567576.Google Scholar
133. Hambidge, KM & Krebs, NF (2007) Zinc deficiency: a special challenge. J Nutr 137, 11011105.Google Scholar
134. Golden, MHN & Golden, BE (1981) Effect of zinc supplementation on the dietary intake, rate of weight gain and energy cost of tissue deposition in children recovering from severe malnutrition. Am J Clin Nutr 34, 900905.Google Scholar
135. Giugliano, R & Millward, DJ (1984) Growth and zinc homeostasis in the severely zinc deficient rat. Br J Nutr 52, 545560.Google Scholar
136. Rossi, L, Migliaccio, S, Corsi, A, et al. (2001) Reduced growth and skeletal changes in zinc-deficient growing rats are due to impaired growth plate activity and inanition. J Nutr 131, 11421146.CrossRefGoogle ScholarPubMed
137. Williams, RB & Mills, CF (1970) The experimental production of zinc deficiency in the rat. Br J Nutr 24, 9891003.Google Scholar
138. Giugliano, R & Millward, DJ (1987) The effects of severe zinc deficiency on protein turnover in muscle and thymus. Br J Nutr 51, 139155.Google Scholar
139. Browning, JD, MacDonald, RS, Thornton, WH, et al. (1998) Reduced food intake in zinc deficient rats is normalized by megestrol acetate but not by insulin-like growth factor-I. J Nutr 128, 136142.CrossRefGoogle Scholar
140. MacDonald, RS, Wollard-Biddle, LC, Browning, JD, et al. (1998) Zinc deprivation of murine 3T3 cells by use of diethylenetrinitrilopentaacetate impairs DNA synthesis upon stimulation with insulin-like growth factor-I (IGF-I). J Nutr 128, 16001605.Google Scholar
141. MacDonald, RS (2000) The role of zinc in growth and cell proliferation. J Nutr 130, 1500S1508S.Google Scholar
142. Riesco, G, Taurog, A, Larsen, R, et al. (1977) Acute and chronic responses to iodine deficiency in rats. Endocrinology 100, 303313.CrossRefGoogle ScholarPubMed
143. Moreno-Reyes, R, Egrise, D, Boelaert, M, et al. (2006) Iodine deficiency mitigates growth retardation and osteopenia in selenium-deficient rats. J Nutr 136, 595600.Google Scholar
144. Ren, FL, Guo, X, Zhang, RJ, et al. (2007) Effects of selenium and iodine deficiency on bone, cartilage growth plate and chondrocyte differentiation in two generations of rats. Osteoarthritis Cartilage 15, 11711177.Google Scholar
145. Spiegel, C, Bestetti, GE, Rossi, GL, et al. (1993) Normal circulating triiodothyronine concentrations are maintained despite severe hypothyroidism in growing pigs fed rapeseed presscake meal. J Nutr 123, 15541561.Google Scholar
146. Tata, JR & Widnell, CC (1966) Ribonucleic acid synthesis during the early action of thyroid hormones. Biochem J 98, 604620.Google Scholar
147. Brown, JG, Bates, PC, Holliday, MA, et al. (1981) Thyroid hormones and muscle protein turnover: the effect of thyroid-hormone deficiency and replacement in thyroidectomized and hypophysectomized rats. Biochem J 194, 771782.CrossRefGoogle ScholarPubMed
148. Brown, JG & Millward, DJ (1983) Dose response of protein turnover in rat skeletal muscle to triiodothyronine treatment. Biochim Biophys Acta 757, 182190.Google Scholar
149. O’Connell, JM, Dibley, MJ, Sierra, J, et al. (1989) Growth of vegetarian children: The Farm Study. Pediatrics 84, 475481.Google Scholar
150. Dwyer, JT, Andrew, EM & Berkey, C (1983) Growth in ‘new’ vegetarian preschool children using the Jenss–Bayley curve fitting technique. Am J Clin Nutr 37, 815827.Google Scholar
151. Dagnelie, PC, van Dusseldorp, M, van Staveren, WA, et al. (1994) Effects of macrobiotic diets on linear growth in infants and children until 10 years of age. Eur J Clin Nutr 48, Suppl. 1, S103S111.Google Scholar
152. Van Dusseldorp, M, Arts, ICW, Bergsma, JS, et al. (1996) Catch-up growth in children fed a macrobiotic diet in early childhood. J Nutr 126, 29772983.Google Scholar
153. Allen, LH & Gillespie, SR (2001) What Works? A Review of the Efficacy and Effectiveness of Nutrition Interventions. Manila: Asian Development Bank; Geneva: ACC/SCN.Google Scholar
154. Martorell, R & Klein, RE (1980) Food supplement and growth rates in preschool children. Nutr Rep Int 21, 447454.Google Scholar
155. Lutter, CK, Mora, JO, Habicht, J-P, et al. (1990) Age-specific responsiveness of weight and length to nutritional supplementation. Am J Clin Nutr 51, 359364.Google Scholar
156. Walker, SP, Powell, CA, Grantham-McGregor, SM, et al. (1991) Nutritional supplementation, psychosocial stimulation, and growth of stunted children: the Jamaican study. Am J Clin Nutr 54, 642648.Google Scholar
157. Hegsted, DM (1972) Deprivation syndrome or protein–calorie malnutrition. Nutr Rev 30, 5154.Google Scholar
158. Gopalan, C, Swaminathan, MC, Kumari, VK, et al. (1973) Effect of calorie supplementation on growth of undernourished children. Am J Clin Nutr 26, 563566.Google Scholar
159. Habicht, JP, Martorell, R & Rivera, JA (1995) Nutritional impact of supplementation in the INCAP longitudinal study: analytic strategies and inferences. J Nutr 125, 1042S1050S.Google Scholar
160. Martorell, R, Habicht, JP & Rivera, JA (1995) History and design of the INCAP longitudinal study (1969–77) and its follow-up (1988–89). J Nutr 125, 1027S1041S.Google Scholar
161. Allen, LH (1995) Malnutrition and human function: a comparison of conclusions from the INCAP and nutrition CRSP studies. J Nutr 125, 1119S1126S.Google Scholar
162. Krebs, NF, Mazariegos, M, Chomba, E, et al. (2012) Randomized controlled trial of meat compared with multimicronutrient-fortified cereal in infants and toddlers with high stunting rates in diverse settings. Am J Clin Nutr 96, 840847.Google Scholar
163. Malcolm, LA (1970) Growth retardation in a New Guinea boarding school and its response to supplementary feeding. Br J Nutr 24, 297305.Google Scholar
164. Lampl, M, Johnston, FE & Malcolm, LA (1978) The effects of protein supplementation on the growth and skeletal maturation of New Guinean school children. Ann Hum Biol 5, 219227.Google Scholar
165. Malcolm, LA (1970) Growth and Development in New Guinea: A Study of the Bundi People of the Madang District, Monograph Series No. 1. Madang: Institute of Human Biology of Papua and New Guinea.Google Scholar
166. Garn, SM (1972) Growth and development in New Guinea – A study of the Bundi people of the Madang district. By L. A. Malcolm. 105 pp., figures, tables, appendices, bibliography. Monograph Series No. 1, Institute of Human Biology of Papua and New Guinea, Madang. 1970 [Book review]. Am J Phys Anthropol 37, 157159.Google Scholar
167. World Health Organization (2007) Protein and Amino Acid Requirements in Human Nutrition. Report of a Joint WHO/FAO/UNU Expert Consultation. WHO Technical Report Series no. 935. Geneva: WHO.Google Scholar
168. Millward, DJ & Jackson, AA (2003) Protein/energy ratios of current diets in developed and developing countries compared with a safe protein/energy ratio: implications for recommended protein and amino acid intakes. Public Health Nutr 7, 387405.Google Scholar
169. Heinig, MJ, Nommsen, LA, Peerson, JM, et al. (1993) Energy and protein intakes of breast-fed and formula-fed infants during the first year of life and their association with growth velocity: the DARLING Study. Am J Clin Nutr 58, 152161.Google Scholar
170. Räiha, NC, Fazzolari-Nesci, A, Cajozzo, C, et al. (2002) Whey predominant, whey modified infant formula with protein/energy ratio of 1.8 g/100 kcal: adequate and safe for term infants from birth to four months. J Pediatr Gastroenterol Nutr 35, 275281.Google Scholar
171. EFSA Panel on Dietetic Products, Nutrition and Allergies (2014) Scientific opinion on the essential composition of infant and follow-on formulae. EFSA J 12, 3760.Google Scholar
172. Macé, K, Steenhout, P, Klassen, P, et al. (2006) Protein quality and quantity in cow’s milk-based formula for healthy term infants: past, present and future. Nestlé Nutr Workshop Ser Pediatr Program 58, 189205.Google Scholar
173. Hoppe, C, Molgaard, C, Thomsen, BL, et al. (2004) Protein intake at 9 mo of age is associated with body size but not with body fat in 10-y-old Danish children. Am J Clin Nutr 79, 494501.Google Scholar
174. Hörnell, A, Lagström, H, Lande, B, et al. (2013) Protein intake from 0 to 18 years of age and its relation to health: a systematic literature review for the 5th Nordic Nutrition Recommendations. Food Nutr Res 2013, 57.Google Scholar
175. Hoppe, C, Udam, TR, Lauritzen, L, et al. (2004) Animal protein intake, serum insulin-like growth factor I, and growth in healthy 2.5-y-old Danish children. Am J Clin Nutr 80, 447452.Google Scholar
176. Swaminathan, S, Vaz, M & Kurpad, AV (2012) Protein intakes in India. Br J Nutr 108, S50S58.Google Scholar
177. Ghosh, S, Suri, D & Uauy, R (2012) Assessment of protein adequacy in developing countries: quality matters. Br J Nutr 108, S77S87.Google Scholar
178. Mack, PB, Vose, GP, Kinard, CL, et al. (1962) Effects of lysine-supplemented diets on growth and skeletal density of preadolescent children. Am J Clin Nutr 11, 255262.Google Scholar
179. Civitelli, R, Villareal, DT, Agnusdei, D, et al. (1992) Dietary l-lysine and calcium metabolism in humans. Nutrition 8, 400405.Google Scholar
180. Graham, GG, Lembcke, J & Morales, E (1990) Quality-protein maize as the sole source of dietary protein and fat for rapidly growing young children. Pediatrics 85, 8591.Google Scholar
181. Nilupa, S, Gunaratna, H, De Groote, P, et al. (2010) A meta-analysis of community-based studies on quality protein maize. Food Policy 35, 202210.Google Scholar
182. Beaton, GH, Calloway, D & Murphy, SP (1992) Estimated protein intakes of toddlers: predicted prevalence of inadequate intakes in village populations in Egypt, Kenya, and Mexico. Am J Clin Nutr 55, 902911.Google Scholar
183. de Benoist, B, Darnton-Hill, I, Davidsson, L, et al. (2007) Conclusions of the Joint WHO/UNICEF/IAEA/IZiNCG Interagency Meeting on Zinc Status Indicators. Food Nutr Bull 28, S480S484.Google Scholar
184. Wessells, KR & Brown, KH (2012) Estimating the global prevalence of zinc deficiency: results based on zinc availability in national food supplies and the prevalence of stunting. PLOS ONE 7, e50568.Google Scholar
185. Bhutta, ZA, Das, JK, Rizvi, A, et al. (2013) Evidence-based interventions for improvement of maternal and child nutrition: what can be done and at what cost? Lancet 382, 452477.Google Scholar
186. Yakoob, MY, Theodoratou, E, Jabeen, A, et al. (2011) Preventive zinc supplementation in developing countries: impact on mortality and morbidity due to diarrhea, pneumonia and malaria. BMC Public Health 11, Suppl. 3, S23.Google Scholar
187. Brown, KH, Peerson, JM & Allen, LH (1997) Effect of zinc supplementation on children’s growth: a meta-analysis of intervention trials. Bibl Nutr Dieta 54, 7683.Google Scholar
188. Imdad, A & Bhutta, ZA (2011) Effect of preventive zinc supplementation on linear growth in children under 5 years of age in developing countries: a meta-analysis of studies for input to the lives saved tool. BMC Public Health 11, Suppl. 3, S22.Google Scholar
189. Mayo-Wilson, E, Junior, JA, Imdad, A, et al. (2014) Zinc supplementation for preventing mortality, morbidity, and growth failure in children aged 6 months to 12 years of age. Cochrane Database of Systematic Reviews, Issue 5. Art. No. CD009384. doi: 10.1002/14651858.CD009384.pub2 Google Scholar
190. Phuka, JC, Maleta, K, Thakwalakwa, C, et al. (2008) Complementary feeding with fortified spread and the incidence of severe stunting among 6- to-18-month-old rural Malawians. Arch Pediatr Adolesc Med 162, 619626.Google Scholar
191. Mangani, C, Maleta, K, Phuka, J, et al. (2015) Effect of complementary feeding with lipid based nutrient supplements and corn–soy blend on the incidence of stunting and linear growth among 6- to 18-month-old infants and children in rural Malawi. Matern Child Nutr 11, Suppl. 4, 132143.Google Scholar
192. Ashorn, P, Alho, L, Ashorn, U, et al. (2015) Supplementation of maternal diets during pregnancy and for 6 months postpartum and infant diets thereafter with small-quantity lipid-based nutrient supplements does not promote child growth by 18 months of age in rural Malawi: a randomized controlled trial. J Nutr 145, 13451353.Google Scholar
193. Adu-Afarwuah, S, Lartey, A, Brown, KH, et al. (2007) Randomized comparison of 3 types of micronutrient supplements for home fortification of complementary foods in Ghana: effects on growth and motor development. Am J Clin Nutr 86, 412420.Google Scholar
194. Miell, JP, Zini, M, Quin, JD, et al. (1994) Reversible effects of cessation and recommencement of thyroxine treatment on insulin-like growth factors (IGFs) and IGF-binding proteins in patients with total thyroidectomy. J Clin Endocrinol Metab 79, 15071512.Google Scholar
195. Iglesias, P, Bayon, C, Mendez, J, et al. (2001) Serum insulin-like growth factor type 1, insulin-like growth factor-binding protein-1, and insulin-like growth factor-binding protein-3 concentrations in patients with thyroid dysfunction. Thyroid 11, 10431048.Google Scholar
196. Zhou, SJ, Anderson, AJ, Gibson, RA, et al. (2013) Effect of iodine supplementation in pregnancy on child development and other clinical outcomes: a systematic review of randomized controlled trials. Am J Clin Nutr 98, 12411254.Google Scholar
197. Hernandez-Cassis, C, Cure-Cure, C & Lopez-Jaramillo, P (1995) Effect of thyroid replacement therapy on the stature of Colombian children with minimal thyroid dysfunction. Eur J Clin Invest 25, 454456.Google Scholar
198. Centers for Disease Control and Prevention (2016) International Micronutrient Malnutrition Prevention and Control (IMMPaCt). http://www.cdc.gov/immpact/about/index.html (accessed August 2016).Google Scholar
199. United Nations Children’s Fund, World Health Organization & United Nations University (1999) Composition of a multi-micronutrient supplement to be used in pilot programmes among pregnant women in developing countries: report of a United Nations Children’s Fund (UNICEF), World Health Organization (WHO) and United Nations University workshop UNICEF. http://www.who.int/iris/handle/10665/75358 (accessed August 2016).Google Scholar
200. Haider, BA & Bhutta, ZA (2015) Multiple-micronutrient supplementation for women during pregnancy. Cochrane Database of Systematic Reviews, Issue 11. Art. No. CD004905. doi: 10.1002/14651858.CD004905.pub4.Google Scholar
201. Rivera, JA, Gonzalez-Cossio, T, Flores, M, et al. (2001) Multiple micronutrient supplementation increases the growth of Mexican infants. Am J Clin Nutr 74, 657663.Google Scholar
202. De-Regil, LM, Suchdev, PS, Vist, GE, et al. (2011) Home fortification of foods with multiple micronutrient powders for health and nutrition in children under two years of age. Cochrane Database of Systematic Reviews, Issue 9. Art. No. CD008959. doi: 10.1002/14651858.CD008959.pub2.Google Scholar
203. Salam, RA, MacPhail, C, Das, JK, et al. (2013) Effectiveness of micronutrient powders (MNP) in women and children. BMC Public Health 13, Suppl. 3, S22.Google Scholar
204. Arimond, M, Zeilani, M, Jungjohann, S, et al. (2015) Considerations in developing lipid-based nutrient supplements for prevention of undernutrition: experience from the International Lipid-Based Nutrient Supplements (iLiNS) Project. Matern Child Nutr 11, 3161.Google Scholar
205. International Lipid-Based Nutrient Supplements Project (2016) iLiNS project. http://www.ilins.org/ (accessed October 2016).Google Scholar
206. Arnold, BF, Null, C, Luby, SP, et al. (2013) Cluster-randomised controlled trials of individual and combined water, sanitation, hygiene and nutritional interventions in rural Bangladesh and Kenya: the WASH Benefits study design and rationale. BMJ Open 3, e003476.Google Scholar
207. The Sanitation Hygiene Infant Nutrition Efficacy (SHINE) Trial Team (2015) The Sanitation Hygiene Infant Nutrition Efficacy (SHINE) Trial: rationale, design, and methods. Clin Infect Dis 61, Suppl. 7, S685S702.Google Scholar
208. Neumann, CG, Harris, DM & Rogers, LM (2002) Contribution of animal source foods in improving diet quality and function in children in the developing world. Nutr Res 22, 193220.Google Scholar
209. Allen, LH & Dror, DK (2011) Effects of animal source foods, with emphasis on milk, in the diet of children in low-income countries. In Milk and Milk Products in Human Nutrition, pp. 113130 [RA Clemens, O Hernell and KF Michaelsen, editors]. Basel: Nestec Ltd.Google Scholar
210. Neumann, CG, Bwibo, NO, Murphy, SP, et al. (2003) Animal source foods improve dietary quality, micronutrient status, growth and cognitive function in Kenyan school children: background, study design and baseline findings. J Nutr 133, 3941S3949S.Google Scholar
211. Neumann, CG, Murphy, SP, Gewa, C, et al. (2007) Meat supplementation improves growth, cognitive, and behavioral outcomes in Kenyan children. J Nutr 137, 11191123.Google Scholar
212. Pollock, JI (2006) Two controlled trials of supplementary feeding of British school children in the 1920s. J R Soc Med 99, 323327.Google Scholar
213. Atkins, PJ (2005) The Milk in Schools Scheme, 1934–45: ‘nationalization’ and resistance. Hist Educ 34, 121.Google Scholar
214. Hoppe, C, Mølgaard, C & Michaelsen, KF (2006) Cow’s milk and linear growth in industrialized and developing countries. Annu Rev Nutr 26, 131173.Google Scholar
215. Takahashi, E (1971) Geographic distribution of human stature and environmental factors – an ecologic study. J Anthropol Soc Nippon 76, 259285.Google Scholar
216. Takahashi, E (1984) Secular trend in milk consumption and growth in Japan. Hum Biol 56, 427437.Google Scholar
217. Bogin, B (1988) Patterns of Human Growth. New York: Cambridge University Press.Google Scholar
218. Bogin, B (1998) Milk and human development: an essay on the “milk hypothesis”. Antropol Portuguesa 15, 2336.Google Scholar
219. Wiley, AS (2005) Does milk make children grow? Relationships between milk consumption and height in NHANES 1999–2002. Am J Hum Biol 17, 425441.Google Scholar
220. Black, RE, Williams, SM, Jones, IE, et al. (2002) Children who avoid drinking cow milk have low dietary Ca intakes and poor bone health. Am J Clin Nutr 76, 675680.Google Scholar
221. Romero, IG, Mallick, CB, Liebert, A, et al. (2012) Herders of Indian and European cattle share their predominant allele for lactase persistence. Mol Biol Evol 29, 249260.Google Scholar
222. Melnik, BC, John, SM & Schmitz, G (2013) Milk is not just food but most likely a genetic transfection system activating mTORC1 signalling for postnatal growth. Nutr J 12, 103.Google Scholar
223. Hoppe, C, Mølgaard, C, Juul, A, et al. (2004) High intakes of skimmed milk, but not meat, increase serum IGF-I and IGFBP-3 in eight-year-old boys. Eur J Clin Nutr 58, 12111216.Google Scholar
224. Hoppe, C, Mølgaard, C, Vaag, A, et al. (2005) High intakes of milk, but not meat, increases s-insulin and insulin resistance in 8-year-old boys. Eur J Clin Nutr 59, 393398.Google Scholar
225. Hegsted, DM (2001) Fractures, calcium, and the modern diet. Am J Clin Nutr 74, 571573.Google Scholar
226. Ruel, MT (2003) Milk intake is associated with better growth in Latin America: evidence from the Demographic and Health Surveys. J FASEB 17, A1199.Google Scholar
227. Rutishauser, IHE & Whitehead, RG (1969) Nutritional status assessed by biological tests. Br J Nutr 23, 113.Google Scholar
228. Martorell, R, Schroeder, DG, Rivera, JA, et al. (1995) Patterns of linear growth in rural Guatemalan adolescents and children. J Nutr 125, Suppl. 4, 1060S1067S.Google Scholar
229. World Health Organization, United Nations Children’s Fund (UNICEF) & International Council for the Control of Iodine Deficiency Disorders (ICCIDD) (2007) Assessment of Iodine Deficiency Disorders and Monitoring their Elimination, 3rd ed. Geneva: WHO.Google Scholar
230. Pharoah, PO & Connolly, KJ (1987) A controlled trial of iodinated oil for the prevention of endemic cretinism: a long-term follow-up. Int J Epidemiol 16, 6873.Google Scholar
231. Stephensen, CB (1999) Burden of infection on growth failure. J Nutr 129, 534S538S.Google Scholar
232. Guerrant, RL, Oriá, RB, Moore, SR, et al. (2008) Malnutrition as an enteric infectious disease with long-term effects on child development. Nutr Rev 66, 487505.Google Scholar
233. Kalantar-Zadeh, K, Block, G, McAllister, CJ, et al. (2004) Appetite and inflammation, nutrition, anemia, and clinical outcome in hemodialysis patients. Am J Clin Nutr 80, 299307.Google Scholar
234. McMillan, DC, Wigmore, SJ, Fearon, KCH, et al. (1999) A prospective randomized study of megestrol acetate and ibuprofen in gastrointestinal cancer patients with weight loss. Br J Cancer 79, 495500.Google Scholar
235. Benjamin, DK Jr, Miller, WC, Benjamin, DK, et al. (2003) A comparison of height and weight velocity as part of the composite endpoint in pediatric HIV. AIDS 17, 23312336.Google Scholar
236. Arpadi, SM (2005) Growth failure in HIV-infected children infected children. WHO Consultation on Nutrition and HIV/AIDS in Africa: Evidence, lessons and recommendations for action. http://www.who.int/nutrition/topics/PN4_Growthfailure_Durban.pdf?ua=1 (accessed October 2016).Google Scholar
237. Henderson, RA, Saavedra, JM, Perman, JA, et al. (1994) Effects of enteral tube feeding on growth of children with symptomatic human immunodeficiency virus infection. J Pediatr Gastroenterol Nutr 18, 429434.Google Scholar
238. Clarick, RH, Hanekom, WA, Yogev, R, et al. (1997) Megestrol acetate treatment of growth failure in children with human immunodeficiency virus. Pediatrics 99, 354357.Google Scholar
239. Aeberli, I, Molinari, L, Spinas, G, et al. (2006) Dietary intakes of fat and antioxidant vitamins are predictors of subclinical inflammation in overweight Swiss children. Am J Clin Nutr 84, 748755.Google Scholar
240. Kruger, HS, Pretorius, R & Schutte, AE (2010) Stunting, adiposity, and low-grade inflammation in African adolescents from a township high school. Nutrition 26, 9099.Google Scholar
241. Checkley, W, Buckley, G, Gilman, RH, et al. (2008) Multi-country analysis of the effects of diarrhoea on childhood stunting. Int J Epidemiol 37, 816830.Google Scholar
242. Bhutta, ZA, Ahmed, T, Black, RE, et al. (2008) Maternal and child undernutrition 3: what works? Interventions for maternal and child undernutrition and survival. Lancet 371, 417440.Google Scholar
243. Solomons, NW, Mazariegos, M, Brown, KH, et al. (1993) The underprivileged, developing country child: environmental contamination and growth failure revisited. Nutr Rev 51, 327332.Google Scholar
244. Richard, SA, Black, RE, Gilman, RH, et al. (2013) Diarrhea in early childhood: short-term association with weight and long-term association with length. Am J Epidemiol 178, 11291138.Google Scholar
245. Lunn, PG, Northrop-Clewes, CA & Downes, RM (1991) Intestinal permeability, mucosal injury, and growth faltering in Gambian infants. Lancet 338, 907910.Google Scholar
246. Campbell, DI, Elia, M & Lunn, PG (2003) Growth faltering in rural Gambian infants is associated with impaired small intestinal barrier function, leading to endotoxemia and systemic inflammation. J Nutr 133, 13321338.Google Scholar
247. Campbell, DI, Murch, SH, Elia, M, et al. (2003) Chronic T cell-mediated enteropathy in rural West African children: relationship with nutritional status and small bowel function. Pediatr Res 54, 306311.Google Scholar
248. Kotloff, KL, Nataro, JP, Blackwelder, WC, et al. (2013) Burden and aetiology of diarrhoeal disease in infants and young children in developing countries (the Global Enteric Multicenter Study, GEMS): a prospective, case–control study. Lancet 382, 209222.Google Scholar
249. Humphrey, JH (2009) Child undernutrition, tropical enteropathy, toilets, and handwashing. Lancet 374, 10321035.Google Scholar
250. Korpe, PS & Petri, WA Jr (2012) Environmental enteropathy: critical implications of a poorly understood condition. Trends Mol Med 18, 328336.Google Scholar
251. Keusch, GT, Rosenberg, IH, Denno, DM, et al. (2013) Implications of acquired environmental enteric dysfunction for growth and stunting in infants and children living in low- and middle-income countries. Food Nutr Bull 34, 357364.Google Scholar
252. Crane, RJ, Jones, KDJ & Berkley, JA (2015) Environmental enteric dysfunction: an overview. Food Nutr Bull 36, S76S87.Google Scholar
253. Gough, EK, Stephens, DA, Moodie, EE, et al. (2015) Linear growth faltering in infants is associated with Acidaminococcus sp. and community-level changes in the gut microbiota. Microbiome 3, 24.Google Scholar
254. Smith, LE, Prendergast, AJ, Turner, PC, et al. (2015) Sanitation Hygiene Infant Nutrition Efficacy (SHINE) Trial Team. The potential role of mycotoxins as a contributor to stunting in the SHINE Trial. Clin Infect Dis 61, Suppl. 7, S733S737.Google Scholar
255. Bennett, JW & Klich, M (2003) Mycotoxins. Clin Microbiol Rev 16, 497516.Google Scholar
256. Hendrickse, RG, Coulter, JB & Lamplugh, SM (1982) Aflatoxins and kwashiorkor: a study in Sudanese children. Br Med J (Clin Res Ed) 285, 843846.Google Scholar
257. Golden, MHN (1998) Oedematous malnutrition. Br Med Bull 54, 433444.Google Scholar
258. Lin, A, Arnold, BF, Afreen, S, et al. (2013) Household environmental conditions are associated with enteropathy and impaired growth in rural Bangladesh. Am J Trop Med Hyg 89, 130137.Google Scholar
259. Dewey, KG & Adu-Afarwuah, S (2008) Systematic review of the efficacy and effectiveness of complementary feeding interventions in developing countries. Matern Child Nutr 4, Suppl. 1, 2485.Google Scholar
260. Mbuya, MNN, Tavengwa, NV, Stoltzfus, RJ, et al. (2015) Design of an intervention to minimize ingestion of fecal microbes by young children in rural Zimbabwe. Clin Infect Dis 61, Suppl. 7, S703S709.Google Scholar
261. Desai, A, Smith, LE, Mbuya, MNN, et al. (2015) The SHINE Trial infant feeding intervention: pilot study of effects on maternal learning and infant diet quality in rural Zimbabwe. Clin Infect Dis 61, Suppl. 7, S710S715.Google Scholar
Figure 0

Fig. 1 The stunting syndrome. Modified and simplified from Prendergast & Humphrey(10). The shaded boxes indicate adverse influences while the unshaded boxes indicate outcomes. Stunting is identified as a cyclical process, often starting in utero, connecting maternal nutrition to an intergenerational cycle of growth failure transmitted across generations through the mother(11). High rates are apparent for at least the first 2 years of postnatal life, hence the identification of the critical window of the first 1000 d(6) but growth continues to falter in poor environments with no indication of a levelling off(5), resulting in school children and adults of short stature. Mothers with short stature and especially teenagers are more likely to have low-birth-weight babies who are subsequently more likely to have growth failure during childhood(12). Potential mechanisms explaining intergenerational effects on linear growth are shared genetic characteristics, epigenetic effects, programming of metabolic changes, the mechanics of a reduced space for fetal growth(6) and sociocultural factors such as the intergenerational transmission of poverty and deprivation(13). Multiple environmental influences contribute to impaired growth including poor maternal and child nutrition throughout the cycle, inadequate breast-feeding and inappropriate complementary feeding(14) together with infectious and inflammatory insults(15). These interactions are mutually reinforcing through infection exacerbating any malnutrition, because of appetite suppression and reduced food intake, and any malabsorption reducing nutrient intake, while malnutrition reduces immune defence systems, thereby worsening the adverse influence of infections. The multiple pathological changes marked by linear growth retardation in early life are associated with increased morbidity and mortality, reduced physical, neurodevelopmental and economic capacity and an elevated risk of metabolic disease into adulthood(10). IQ, intelligence quotient; SGA, small for gestational age; prem, prematurity; HAZ, height-for-age Z score.

Figure 1

Fig. 2 Physiological coordination of whole-body growth. The hierarchy of height-growth control involves bone length growth, regulating skeletal muscle growth, which influences whole-body energy expenditure, consequent food intake and the size of the visceral organ mass. See Millward(27) for details, including the molecular basis of the muscle growth response to passive stretch by bone.

Figure 2

Fig. 3 Stunting as a predominantly inflammatory disease resulting from poor hygiene and environmental enteric dysfunction. The pathway between an unsanitary environment, dietary mycotoxins and other potential pathogens, an abnormal intestinal microbiota and stunting includes intestinal inflammation and pathological changes to the GI mucosa. This results in a failure of barrier function allowing translocation of pathogens and endotoxins resulting in a systemic inflammatory response which inhibits bone growth. In addition, nutrient malabsorption occurs exacerbating any dietary insufficiency, worsening malnutrition and increasing susceptibility to infection through its adverse effect on the immune system. This also contributes to the bone growth inhibition. GH, growth hormone; IGF, insulin-like growth factor; IGFBP, insulin-like growth factor binding protein. Adapted from Humphrey(249), Korpe & Petri(250), Keusch et al.(251) and Crane et al.(252).

Figure 3

Fig. 4 Role of nutrition and inflammation in the regulation of endochondral ossification. During the three developmental growth phases of the infancy, childhood and puberty (ICP) model of Karlberg(20), the primary endocrine activators of growth are insulin (infancy), growth hormone (childhood) and the sex steroids (puberty) (although current evidence suggests that growth hormone may also be involved in the infancy stage). These act on the paracrine/autocrine system within the growth plate which mediate endochondral ossification. However, a suitable nutritional anabolic drive is necessary for this process to occur involving both type I nutrients such as iodine (to enable adequate thyroid hormone production) and type II nutrients of which amino acids and zinc are particularly important in stimulating the endocrine/paracrine system and in having direct regulatory influences, although this is poorly understood at the molecular level. Milk also appears to have a specific influence not observed with other animal-source foods. Elevated levels of glucocorticoids, inhibitory fibroblast growth factors (for example, FGF21), pro inflammatory cytokines, especially TNFα, IL-1β, IL-6 and other inflammatory mediators associated with infection and inflammation and environmental enteric dysfunction as in Fig. 3, block the nutritional anabolic drive and inhibit endochondral ossification. How these interactions between nutrition and infection occur at the cellular and molecular level is poorly understood. IGF, insulin-like growth factor; IGFBP, insulin-like growth factor binding protein; T3, triiodothyronine; 1,25(OH)2D, 1,25-dihydroxyvitamin D; IHH, Indian hedgehog; PTHrP, parathyroid-hormone-related protein; BMP, bone morphogenetic protein; WNT, wingless/integrated protein; VEGF, vascular endothelial growth factor; EED, environmental enteric dysfunction.