Hostname: page-component-848d4c4894-2pzkn Total loading time: 0 Render date: 2024-04-30T14:34:02.595Z Has data issue: false hasContentIssue false

Exploring potential mechanisms for zinc deficiency to impact in autism spectrum disorder: a narrative review

Published online by Cambridge University Press:  20 September 2023

M.V. Conti*
Affiliation:
Laboratory of Dietetics and Clinical Nutrition, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, Pavia, Italy.
S. Santero
Affiliation:
Laboratory of Dietetics and Clinical Nutrition, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, Pavia, Italy.
A. Luzzi
Affiliation:
Clinical Nutrition Unit, General Medicine, ICS Maugeri IRCCS, Pavia, Italy Post Graduate Course in Food Science and Human Nutrition, Università Statale di Milano, Milan, Italy
H. Cena
Affiliation:
Laboratory of Dietetics and Clinical Nutrition, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, Pavia, Italy. Clinical Nutrition Unit, General Medicine, ICS Maugeri IRCCS, Pavia, Italy
*
*Corresponding author: M.V. Conti, email: mariavittoria.conti@unipv.it
Rights & Permissions [Opens in a new window]

Abstract

Autism spectrum disorder (ASD) is a heterogeneous and complex group of life-long neurodevelopmental disorders. How this clinical condition impacts an individual’s intellectual, social and emotional capacities, contributing to alterations in the proprioceptive and sensory systems and increasing their selective attitude towards food, is well described in the literature. This complex condition or status exposes individuals with ASD to an increased risk of developing overweight, obesity and non-communicable diseases compared with the neurotypical population. Moreover, individuals with ASD are characterised by higher levels of inflammation, oxidative stress markers and intestinal dysbiosis. All these clinical features may also appear in zinc deficiency (ZD) condition. In fact, zinc is an essential micronutrient for human health, serving as a structural, catalytic and regulatory component in numerous physiological processes. The aim of this narrative review is to explore role of ZD in ASD. Factors affecting zinc absorption, excretion and dietary intake in this vulnerable population are taken into consideration. Starting from this manuscript, the authors encourage future research to investigate the role of ZD in ASD. The perspective is to potentially find another missing piece in the ‘ASD clinical puzzle picture’ to improve the health status of these individuals.

Type
Review Article
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an Open Access article, distributed under the terms of the Creative Commons Attribution licence (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted re-use, distribution and reproduction, provided the original article is properly cited.
Copyright
© The Author(s), 2023. Published by Cambridge University Press on behalf of The Nutrition Society

Introduction

Zinc (Zn) is a redox neutral IIB group metal(Reference Kambe, Tsuji and Hashimoto1) and an essential micronutrient for human health. In fact, it is the second most abundant divalent cation after calcium and serves as a structural, catalytic and regulatory component(Reference Chasapis, Ntoupa and Spiliopoulou2) in numerous physiological processes. It is necessary for the structure of over 2000 transcription factors(Reference Chasapis, Ntoupa and Spiliopoulou2), and more than 300 enzymes depend on it for their functioning(Reference Skalny, Aschner and Tinkov3).

Moreover, it is able to modulate numerous intracellular signalling pathways, as well as influencing the progression of the cell cycle itself, in addition to fulfilling its antioxidant and anti-inflammatory roles(Reference Kambe, Tsuji and Hashimoto1Reference Wessels, Fischer and Rink4).

Despite its physiological centrality, the mineral content in the human body is very low, at just 2–3 g. Approximately 95% of its content is intracellular, primarily located in muscles, followed by bones, brain, testicles and liver(Reference Chasapis, Ntoupa and Spiliopoulou2,Reference Livingstone5) . Zinc is not stored in the body and undergoes a rapid turnover. Therefore, maintaining adequate dietary intake is necessary to support all the functions mentioned above(Reference Chasapis, Ntoupa and Spiliopoulou2,Reference Livingstone5) .

Therefore, ensuring the appropriate daily dietary intake is essential to ensure an optimal health. This is particularly crucial for ‘vulnerable’ individuals who are at higher risk of not meeting the body’s requirements(Reference Valenzuela-Zamora, Ramírez-Valenzuela and Ramos-Jiménez6Reference Marí-Bauset, Zazpe and Mari-Sanchis8), resulting in the development of Zn deficiency (ZD) and subsequent health implications(Reference Chasapis, Ntoupa and Spiliopoulou2,Reference Skalny, Aschner and Tinkov3,Reference Alsufiani, Alkhanbashi and Laswad9,Reference Kuschner, Eisenberg and Orionzi10) .

The general causes of ZD include inadequate Zn intake, increased Zn requirements, reduced Zn absorption, increased Zn excretion and impaired utilisation(Reference Chasapis, Ntoupa and Spiliopoulou2,Reference Livingstone5,Reference Roohani, Hurrell and Kelishadi11) .

The prevalence of ZD is estimated to be approximately 17% globally(Reference Chasapis, Ntoupa and Spiliopoulou2), and it is more frequently diagnosed in developing countries(Reference Skalny, Aschner and Tinkov3). Nevertheless, ZD is also prevalent in developed countries, including Italy, due to multiple factors such as reduced Zn absorption, gastrointestinal (GI) diseases, ageing and/or the presence of specific pathological conditions(12). Moreover, ZD is shown to be highly prevalent in individuals with autism spectrum disorders (ASD) compared with neurotypical individuals(Reference Alsufiani, Alkhanbashi and Laswad9). This could be attributed to the frequent occurrence of food selectivity and the comorbidities that characterise individuals with ASD.

ASD is a heterogeneous and complex group of neurodevelopmental disorders(13). According to the DSM-V(13), the diagnostic criteria of ASD must involve two dimensions: (A) persistent deficit of social communication and social interaction in multiple contexts; (B) restricted, repetitive patterns of behaviour, interests or activities(13). This dimensional diagnosis must be combined with other specific descriptors that outline the intensity of the disorder, such as: (i) with or without accompanying intellectual impairment; (ii) with or without accompanying language impairment; (iii) associated with a known medical or genetic condition or environmental factor; (iv) associated with another neurodevelopmental, mental or behavioural disorder(13). Additionally, another crucial evaluation criterion is the level of severity and support required, which is described in three levels: level 3 requires very substantial support; level 2 requires substantial support; level 1 requires support(13). Furthermore, approximately 75% of individuals with ASD have other comorbidities that impact their physical and mental state, such as attention-deficit/hyperactivity disorder (ADHD), depressive and anxiety disorders, bipolar disorder, obsessive–compulsive disorder, irritable bowel, inflammatory bowel disease, epilepsy, immune disorders, and sensory and sleep disorders(Reference Sharma, Gonda and Tarazi14).

Moreover, traits such as systematic and neurological inflammation, oxidative stress, gastrointestinal (GI) symptoms, overweight and obesity as well as food selectivity (FS) are frequently observed in individuals with ASD, with a significantly higher prevalence than in the general population(Reference Valenzuela-Zamora, Ramírez-Valenzuela and Ramos-Jiménez6,Reference Kuschner, Eisenberg and Orionzi10,Reference Sauer, Malijauskaite and Meleady15,Reference Bjørklund, Meguid and El-Bana16) . In fact, according to a recent meta-analysis, the prevalence of overweight and obesity are respectively 19·8% and 21·8% in individuals with ASD(Reference Li, Xie and Lei17). Delving into the topic of FS, it is a condition characterised by a marked limitation of the repertoire of foods accepted, exposing the individual to the risk of developing obesity and micronutrient deficiency, including ZD(Reference Valenzuela-Zamora, Ramírez-Valenzuela and Ramos-Jiménez6,Reference Wohlmacher18) . The prevalence of FS in the paediatric ASD population ranges from 22·9% to 69·1%(Reference Valenzuela-Zamora, Ramírez-Valenzuela and Ramos-Jiménez6) and remains an important issue for adolescents and young adults with ASD(Reference Kuschner, Eisenberg and Orionzi10). The health consequences for these individuals varies depending on the severity of food refusal, the limitations in the food repertoire, and the degree of repetitiveness of feeding behaviour (i.e. high-frequency intake of single food)(Reference Valenzuela-Zamora, Ramírez-Valenzuela and Ramos-Jiménez6,Reference Bandini, Anderson and Curtin19) . Therefore, FS, which involves the adoption of an imbalanced dietary pattern, acts as both a risk factor and a maintenance factor for the development of micronutrient deficiencies, including ZD, and overweight or obesity throughout the lifespan of individuals with ASD.

The purpose of this narrative review is to explore the possible connections between ZD and ASD, including the factors that affect Zn absorption, excretion and dietary intake.

The role of Zn in human systems with a focus on individuals with ASD

Zn plays an important role in the development and functioning of the immune, gastrointestinal and nervous systems, all of which are frequently dysregulated in individuals with ASD(Reference Chasapis, Ntoupa and Spiliopoulou2,Reference Skalny, Aschner and Tinkov3,Reference Sauer, Malijauskaite and Meleady15) .

Starting from the immune system, Zn performs immunomodulatory functions by regulating the proliferation and maturation of T and B lymphocytes, natural killer cells and dendritic cells, as well as antibody production, phagocytosis and antigen presentation(Reference Skalny, Aschner and Tinkov3). Therefore, ZD predisposes individuals to immune disruptions and recurrent infections, including intestinal infectious diseases, which are well described in individuals with ASD(Reference Chasapis, Ntoupa and Spiliopoulou2,Reference Valenzuela-Zamora, Ramírez-Valenzuela and Ramos-Jiménez6,Reference Siniscalco, Schultz and Brigida20,Reference Ohashi and Fukada21) .

Focusing on the gut, Zn is involved in its morphological development, microbial composition and function, and barrier maintenance(Reference Sauer, Malijauskaite and Meleady15) due to its essential role in cell turnover and repair systems.

Thus, the negative effects of ZD include dynamic variation in gut microbial composition, increased intestinal permeability (leaky gut), activation of pro-inflammatory pathways, and diarrhoea(Reference Chasapis, Ntoupa and Spiliopoulou2,Reference Sauer, Malijauskaite and Meleady15,Reference Ohashi and Fukada21) , which are common manifestations in individuals with ASD(Reference Valenzuela-Zamora, Ramírez-Valenzuela and Ramos-Jiménez6,Reference Sauer, Malijauskaite and Meleady15) . Mounting evidence strongly supports a positive relationship between the extent of GI symptoms and the severity of ASD symptomatology(Reference Fowlie, Cohen and Ming22), as well as a close association between Zn status and autism severity(Reference Skalny, Aschner and Tinkov3). Considering the central role of the intestine in Zn absorption, as well as the common GI symptoms reported in individuals with ASD, the authors delved into each of those aspects. Several landmark studies from the past few decades have concentrated on the gut microbiota in individuals with ASD, revealing a decreased microbial diversity in this population(Reference Dhaliwal, Orsso and Richard23), along with a significant increase in Clostridioides difficile and Candida albicans, a decrease in Bifidobacterium and Lactobacillus, and low levels of short-chain fatty acids (SCFAs)(Reference Dhaliwal, Orsso and Richard23Reference Srikantha and Mohajeri25). Nevertheless, the description of a comprehensive and distinctive gut microbial pattern in individuals with ASD is still under research. Different studies have shown conflicting results, probably due to heterogeneity of the analysed samples in terms of age, diet, pharmacological treatment, geographic area, comorbidities and the severity of neurobehavioral and gastrointestinal symptoms(Reference Al-Ayadhi, Zayed and Bhat24,Reference Fattorusso, Di Genova and Dell’Isola26) . The alterations of gut microbiota in individuals with ASD are known to have a significant impact on the brain through the microbiota–gut–brain axis(Reference Sauer, Malijauskaite and Meleady15,Reference Fowlie, Cohen and Ming22,Reference Srikantha and Mohajeri25) , exacerbating the typical symptoms of ASD (i.e. limitations in social interactions and communications, and repetitive behaviours)(13,Reference Al-Ayadhi, Zayed and Bhat24) . Furthermore, the increased permeability of the intestinal barrier, frequently co-present with dysbiosis, results in the entry of bacterial metabolites, such as lipopolysaccharide (LPS), into the bloodstream. This triggers a significant increase in neurological and systemic inflammation by altering cytokine levels(Reference Sauer, Malijauskaite and Meleady15,Reference Srikantha and Mohajeri25,Reference Matta, Hill-Yardin and Crack27) . Individuals with ASD have been found to have increased levels of these leaky gut syndrome biomarkers(Reference Al-Ayadhi, Zayed and Bhat24). As it is known, the gut–brain axis operates bidirectionally; therefore, neuroinflammation and alterations in neuronal activities significantly impact the composition of the gut microbiota in individuals with ASD from early childhood(Reference Srikantha and Mohajeri25,Reference Saurman, Margolis and Luna28) .

Dysbiosis and GI symptoms, such as constipation, diarrhoea, bloating, abdominal pain, reflux and vomiting are four times more prevalent in children with ASD compared with neurotypical individuals(Reference Srikantha and Mohajeri25). Furthermore, there is substantial scientific evidence describing the persistence of GI symptom prevalence into adulthood in individuals with ASD(Reference Leader, Barrett and Ferrari29). So, considering that Zn plays a key role in gut health and intestinal microbiota, it may be important to prevent ZD from an early age, so as not to exacerbate dysbiosis and GI symptoms frequently found in individuals with ASD.

Regarding brain activity, Zn plays a key role in neuronal learning and memory processes(Reference Chasapis, Ntoupa and Spiliopoulou2), synaptic plasticity through ProSAP/Shank scaffold, and neurotransmitter metabolism(Reference Skalny, Aschner and Tinkov3), particularly in glutamate. Specifically, Zn2+ is necessary for proper assembly, structuring and functioning of the ProSAP/Shank scaffold(Reference Skalny, Aschner and Tinkov3) protein and is involved in glutamatergic neurotransmission, as Zn2+ forms complexes with glutamate in presynaptic vesicles(Reference Bhandari, Paliwal and Kuhad30,Reference Zheng, Zhu and Qu31) . Numerous studies have demonstrated that impairment of the synaptic ProSAP/Shank scaffold promotes the development of behaviours typically observed in ASD(Reference Skalny, Aschner and Tinkov3,Reference Alsufiani, Alkhanbashi and Laswad9) . Alterations in the balance between excitatory and inhibitory pathways in the nervous system are frequently observed in individuals with ASD(Reference Skalny, Aschner and Tinkov3,Reference Zheng, Zhu and Qu31) .

Therefore, it may be relevant to prevent ZD conditions as early as possible to avoid exacerbating these neurological alterations.

Ultimately, it is necessary to emphasise the role of ZD in inflammation (systemic and neurological) and oxidative stress, both of which are often present in individuals with ASD(Reference Skalny, Aschner and Tinkov3,Reference Sauer, Malijauskaite and Meleady15,Reference Bjørklund, Meguid and El-Bana16) . The literature reports significantly higher plasma and serum levels of proinflammatory cytokines (IL-1β, IL-6, IL-8 and IFN-&x0263;) in individuals with ASD, compared with neurotypical controls(Reference Sauer, Malijauskaite and Meleady15). Moreover, an increase in the levels of IL-1β, IL-6 and IFN-&x0263; in the brain are reported in postmortem ASD studies(Reference Sauer, Malijauskaite and Meleady15).

Individuals with ASD also present elevated levels of reactive oxygen species (ROS) and are considered more vulnerable to oxidative stress due to their reduced glutathione (GSH) reserve capacity and (GSH) antioxidant defence in specific brain regions(Reference Bjørklund, Meguid and El-Bana16). Considering that Zn enhances (GSH) biosynthesis(Reference Chasapis, Ntoupa and Spiliopoulou2), an adequate intake of this mineral could reduce oxidative stress in individuals with ASD.

In conclusion, ASD is frequently characterised by alteration of immune, gastrointestinal and neurological systems, which share inflammation as a common factor. Considering at the same time the potential role of Zn in modulating the previously mentioned systems, it is reasonable that alterations in Zn levels could potentially impact on ASD symptomatology(Reference Valenzuela-Zamora, Ramírez-Valenzuela and Ramos-Jiménez6,Reference Siniscalco, Schultz and Brigida20,Reference Fattorusso, Di Genova and Dell’Isola26,Reference Matta, Hill-Yardin and Crack27) .

The relationships between the role of zinc in metabolism and ASD comorbidities

Considering the underlying pathogenetic mechanisms of ASD metabolic comorbidities and the role of Zn in the same metabolic systems, it is reasonable to assume that ZD status can contribute to the metabolic comorbidities that are present in individuals with ASD leading to bidirectional relationship (graphical abstract), as further explained below.

Scientific research is currently exploring the role of Zn in diabetes mellitus (DM) in terms of glycaemic control, and its role in obesity and metabolic syndrome(Reference Skalny, Aschner and Tinkov3). Low Zn levels in individuals with type 2 DM and obesity were observed(Reference Skalny, Aschner and Tinkov3). A recent systematic review with meta-analysis found that the association between ASD and DM is not currently supported by robust evidence(Reference Cortese, Gabellone and Marzulli32).

Starting from the analysis of carbohydrates metabolism, individuals with ASD often exhibit sugar malabsorption, which may be attributed to a decreased expression of disaccharidases, specifically sodium–glucose transporter 1 (SGLT1) and glucose transporter 5 (GLUT5), in the brush border in the intestinal epithelium(Reference Srikantha and Mohajeri25,Reference Vela, Stark and Socha33) . The remaining sugars in the intestinal lumen can lead to osmotic diarrhoea and can be fermented by the gut microbiota, causing alterations in microbiota composition, small intestinal bacterial overgrowth (SIBO), bloating and flatulence(Reference Srikantha and Mohajeri25). As 30–50% reduction of disaccharidase activity has been observed in cases of chronic ZD(Reference Vela, Stark and Socha33) and given the higher prevalence of ZD in individuals with ASD, there is a possible role for Zn in alleviating frequent intestinal symptoms observed in ASD.

Zn has a central role in glycaemic control: it is involved in synthesis, storage and release of insulin, and it is present in insulin granules(Reference Kambe, Tsuji and Hashimoto1,Reference Olechnowicz, Tinkov and Skalny34,Reference Tamura35) . It influences the maintenance of the GLUT4 transporter and modulates the insulin receptor (INSR) signalling pathway(Reference Wessels, Fischer and Rink4). This aspect is particularly significant considering that youths with ASD have, on average, higher homeostatic model assessment of insulin resistance (HOMA-IR) than neurotypical individuals, regardless of their BMI and pharmacological treatment(Reference Manco, Guerrera and Ravà36).

Moving on to protein metabolism, aside from its role in protein synthesis, protein structure and enzyme catalysis, adequate daily intake of Zn is necessary for proper protein digestion in the gut, due to its role in several digestive enzymes, including carboxypeptidases(Reference Kambe, Tsuji and Hashimoto1), dipeptidase(Reference Berezin, Berezin and Lichtenauer37) and aminopeptidase(Reference Vela, Stark and Socha33). Therefore, the possible presence of a ZD condition might favour bacterial proteolytic pathway (putrefaction), which may be associated with dysbiosis and gastrointestinal symptoms observed in individuals with ASD(Reference Sanctuary, Kain and Angkustsiri38).

Regarding lipid metabolism, individuals with ASD can experience alterations in their blood lipid profile(Reference Gillberg, Fernell and Kočovská39,Reference Luçardo, Monk and Dias40) , on which Zn appears to have an influence(Reference Tamura35,Reference Shi, Zou and Shen41,Reference Santos, Teixeira and Schoenfeld42) . The mechanism behind this is currently not understood, but the effects of Zn levels in terms of both prevention and treatment of cardiovascular diseases (CVDs) have been well described in literature(Reference Tamura35). In fact, a systematic review of prospective cohort studies showed that higher serum Zn levels are associated with a lower risk of CVDs. Furthermore, a recent meta-analysis indicated that low-dose (<25 mg/d) and long-term (>12 weeks) Zn supplementation is associated with improved blood lipid parameters(Reference Santos, Teixeira and Schoenfeld42). This aspect seems to be more important when considering that, according to a recent study by Bishop et al. 2022, about 75% of adults with ASD have at least one CVD risk factor, compared with 40% in neurotypical individuals(Reference Bishop, Charlton and McLean43,Reference Chu, Foster and Samman44) .

Regarding adipose tissue, Zn is involved in several related physiological processes, including leptin synthesis and adipocyte lipid metabolism regulation(Reference Skalny, Aschner and Tinkov3,Reference Olechnowicz, Tinkov and Skalny34) . Given this background, many authors have suggested that Zn status could be associated with the state of adipose tissue in obesity(Reference Skalny, Aschner and Tinkov3). These results are in line with the known higher inflammatory and oxidative state of this tissue in individuals with overweight and obesity(Reference Manna and Jain45). Considering the role of zinc in the anti-inflammatory and antioxidant systems(Reference Skalny, Aschner and Tinkov3,Reference Olechnowicz, Tinkov and Skalny34) , the authors hypothesise that lower Zn levels in adipose tissue might also be present in individuals with ASD. This suggests that screening Zn levels in such individuals may be beneficial for those showing signs of lipid metabolism dysregulation.

In conclusion, considering the possible links discussed thus far, Zn could play a key role in the frequently observed metabolic comorbidities in individuals with ASD. Consequently, the importance of screening Zn levels in individuals with ASD who present with metabolic comorbidities, emerges as a crucial aspect to better manage their clinical condition(Reference Indika, Frye and Rossignol46).

Sensory perception, food selectivity and Zn in individuals with ASD

The role of Zn in sensory perception, specifically taste perception, can be discussed by distinguishing between the mechanisms at the oral and neurological levels.

In the mouth, Zn participates in the maintenance and regeneration of the lingual epithelium and taste buds(Reference Kumbargere Nagraj, George and Shetty47,Reference Nishiguchi, Ohmoto and Koki48) . Moreover, it is necessary for the activity of alkaline phosphatase and gustatin, which are associated with taste and smell alterations when their activity is low(Reference Kumbargere Nagraj, George and Shetty47). Animal studies suggest that Zn influences the expression of some taste receptors and membrane channels, such as bitter taste-sensing type 2 receptors (TAS2Rs) and epithelial Na channel (ENaC)(Reference Ikeda, Sekine and Takao49,Reference Onoda, Hirai and Takao50) , indicating the role of Zn especially in bitter(Reference Fábián, Beck and Fejérdy51,Reference Wang, Zajac and Lei52) and salty taste perception(Reference Braud and Boucher53). The role of Zn bitter taste perception is supported by the involvement of gustatin itself in bitter taste(Reference Fábián, Beck and Fejérdy51) and the finding of a lower frequency of expression of six TAS receptor genes in individuals with hypogeusia compared with healthy controls(Reference Onoda, Hirai and Takao50).

Analysing the neural mechanisms, Zn seems to promote the transmission of information to gustatory nerve fibres and to modulate neuropeptides, such as neuropeptide Y (NPY), and neurotransmitter concentrations in the hypothalamus(Reference Santos54,Reference Baltaci, Mogulkoc and Baltaci55) . Moreover, current literature is exploring the influence of Zn on the levels of leptin, insulin and NPY(Reference Kambe, Tsuji and Hashimoto1,Reference Olechnowicz, Tinkov and Skalny34,Reference Tamura35,Reference Baltaci, Mogulkoc and Baltaci55Reference Komai, Goto and Ohinata57) , in relation to taste perception and taste bud physiology. These peptide hormones are present in saliva, and their respective receptors are expressed in taste cells(Reference Fábián, Beck and Fejérdy51). The multiple roles played by Zn in taste perception are summarised in Table 1.

Table 1. The multiple roles played by Zn in taste perception

Legend. Explanation of the role of Zn in taste perception, distinguishing between the mechanisms put in place at the oral and neurological level.

In light of the evidence described, it is important to consider that alterations in taste sensory perception (e.g. smell, taste and sight) are among the main symptoms of ASD and may persist throughout life(Reference Valenzuela-Zamora, Ramírez-Valenzuela and Ramos-Jiménez6) and can be modulated by several factors, potentially including ZD. The possible co-presence of ZD in individuals with ASD may also be connected to FS, which can lead to micronutrient malnutrition(Reference Valenzuela-Zamora, Ramírez-Valenzuela and Ramos-Jiménez6).

FS itself, often present as a life-long clinical feature in individuals with ASD, can be accompanied by a sensory aversion to food, characterised by a rejection of specific textures, temperatures, flavours, colors and smells(Reference Valenzuela-Zamora, Ramírez-Valenzuela and Ramos-Jiménez6). This attitude leads to the adoption of a diet mainly composed of processed foods with high energy density, rich in sugar and saturated fatty acids, and, consequently, a reduction of dietary diversity(Reference Valenzuela-Zamora, Ramírez-Valenzuela and Ramos-Jiménez6). The typical treatment strategy for FS in individuals with ASD is a personalised, careful and gradual food reintroduction programme using applied behaviour analysis (ABA) techniques(Reference Sarcia58). Although useful, these strategies are often time consuming and difficult to carry out over time for individuals and their families, potentially leading to relapses(Reference Kodak and Piazza59). Due to the potential mechanisms via which ZD could impact in individuals with ASD, described above, it could be relevant to screen ZD in individuals with ASD to design an appropriate and personalised treatment plan. However, there are no current evidence for the role of ZD in FS; therefore, future studies are needed to fill this knowledge gap.

Moreover, taste perception features in individuals with ASD may manifest hyper- or hyposensitivity to food stimuli. In the latter case, individuals may tend towards a greater preference for sweet, salty or spicy foods to achieve an adequate stimulus(Reference Valenzuela-Zamora, Ramírez-Valenzuela and Ramos-Jiménez6).

Concerning Zn supplementation, which has been used since the 1980s(Reference Santos54), a recent systematic review has highlighted that it is the most frequently employed intervention for the prevention and treatment of taste disorders (i.e. ageusia/dysgeusia)(Reference Braud and Boucher53). However, the effectiveness of Zn supplementation and the optimal dosage are still debated and controversial. Moreover, the analysed studies often lacked a control group, and exhibited inconsistencies in terms of intervention duration, sample size, age, sex and comorbidities (often carried out in subjects with cancer or Chronic Kidney Disease). As a result, these studies were generally considered as ‘low quality’ overall(Reference Kumbargere Nagraj, George and Shetty47). Regarding ASD, there are currently no clinical trials exploring the role of Zn in improving taste perception, indicating the need for further research in this area.

The authors therefore emphasise the need for further investigation in this regard, specifically focusing on evaluating the potential role of Zn supplementation in individuals with ASD in relation to alterations in sensory perception.

Factors influencing dietary Zn intake, absorption and excretion in ASD

Inadequate dietary intake of absorbable Zn represents the primary cause of ZD(Reference Roohani, Hurrell and Kelishadi11). This deficiency may result from low dietary intake and/or low bioavailability of dietary Zn.

Causes of ZD under the category of reduced Zn absorption primarily include Inflammatory Bowel Diseases, inherited diseases (i.e. acrodermatitis enteropathica and cystic fibrosis), diarrhoea, unbalanced vegetarian or vegan diet, undernutrition or hidden hunger conditions, eating disorders, alcoholism and exocrine pancreatic insufficiency(Reference Chasapis, Ntoupa and Spiliopoulou2). Causes of ZD under the category of reduced Zn intake are related to food preferences and eating patterns, including conditions such as FS, which is highly prevalent in the ASD population and could lead to suboptimal Zn intake.

Analysing Zn absorption, it is important to point out that the bioavailability of Zn content in food is low, about 20–50%(Reference Baj, Flieger and Flieger60,Reference Maares and Haase61) and it depends on both quantitative and qualitative factors(Reference Maares and Haase61,Reference Shkembi and Huppertz62) . Among the food products, red meat, certain seafood, dairy products, nuts, seeds, legumes and whole-grain cereals are considered good dietary sources of Zn(Reference Shkembi and Huppertz62). Animal products, in particular, are known to provide a more bioavailable source of Zn than plant foods(Reference Maares and Haase61).

Factors with a positive, negative or ‘neutral’ effect on dietary Zn bioavailability are presented in Table 2.

Table 2. Factors with a positive, negative or ‘neutral’ effect on dietary Zn bioavailability

Legend. Factors with a positive, negative or ‘neutral’ effect on dietary Zn bioavailability. The authors developed a +/++/+++ classification based on two aspects: (i) the number of papers in the literature supporting or not supporting the ability of dietary factors to positively/negatively affect Zn absorption; (ii) the magnitude of the resulting effect indicated in the papers on dietary Zn absorption.

Although a low intake of phytate-rich food by individuals with ASD might promote better absorption of dietary Zn since phytates are the most potent inhibitors of Zn absorption(Reference Maares and Haase61,Reference Bel-Serrat, Stammers and Warthon-Medina63,Reference Agnoli, Baroni and Bertini64) , this is not advisable since a reduced consumption of vegetables and legumes increases the risk of micronutrient inadequacy and dysbiosis due to low fibre content(67). Moreover, organic acids, such as malic acid found in fruits, citric acid found in fruits and milk, and lactic acid found in yogurt and fermented foods can improve Zn absorption(Reference Maares and Haase61,Reference Agnoli, Baroni and Bertini64,Reference Melse-Boonstra65) .

Another well-known mechanism that promotes Zn absorption is mediated by the protein content of the diet: free amino acids can bind Zn2+ and be transported with it into the enterocyte(Reference Maares and Haase61). Therefore, eating complete meals with all macronutrients is even more recommendable. This recommendation is also supported by the fact that such individuals with ASD generally accept protein sources, with the exception of legumes(Reference Valenzuela-Zamora, Ramírez-Valenzuela and Ramos-Jiménez6) and fish(Reference Park, Choi and Kim68,Reference Ahumada, Guzmán and Rebolledo69) , which in some cases have a strong smell and taste.

Increased Zn losses represent another category of ZD causes and may result from GI disorders such as diarrhoea, as well as urinary tract disorders including kidney disease and DM(Reference Chasapis, Ntoupa and Spiliopoulou2).

In this regard, the ratio between the amount of absorbed Zn and the fraction of the mineral excreted is crucial, as it affects the cellular content of Zn and its distribution. Over time, due to the initial protective action of homeostatic mechanisms, it also impacts the plasma Zn concentration itself(Reference Chasapis, Ntoupa and Spiliopoulou2,Reference Livingstone5) . In fact, the amount of retained Zn in the human body is highly dependent on its dietary content(Reference Bel-Serrat, Stammers and Warthon-Medina63). Therefore, it is not surprising that Zn is primarily excreted with faeces and secondarily with urine(Reference Maares and Haase61). Severe undernutrition and starvation conditions result in both an increase of urinary Zn losses and a sedentary lifestyle(Reference Bel-Serrat, Stammers and Warthon-Medina63). In fact, a chronic reduction in physical activity level (i.e. sedentary lifestyle) is associated with loss of muscle mass. Increased urinary zinc excretion has been observed in individuals with chronic reduction in physical activity levels(Reference Bel-Serrat, Stammers and Warthon-Medina63), as zinc is well represented within muscle tissue(Reference Chasapis, Ntoupa and Spiliopoulou2). Leading a sedentary lifestyle is common among individuals with ASD(Reference Jones, Downing and Rinehart70), due also to motor comorbidities, so the presence of increased urinary Zn losses is a concrete potential issue in this population, exposing them (along with the other factors previously described) to an increased risk of ZD.

To summarise, Zn absorption is influenced by the composition of food matrix, vegetable food preparation techniques and full meal consumption. On the other hand, Zn elimination is influenced by the possible presence of existing Zn deficiencies or excesses, as well as ongoing catabolic processes, which can be associated with a sedentary lifestyle.

Zn requirements in individuals with ASD. Conclusions and future prospects

Increased Zn requirement constitutes another recognised cause of ZD(Reference Chasapis, Ntoupa and Spiliopoulou2,Reference Roohani, Hurrell and Kelishadi11) . Considering the potential alterations in Zn absorption, excretion or Zn intake previously discussed, the possibility of increased Zn requirements in this population should be considered. It is important to note that the dietary recommendations provided by national and international organisations are intended for the healthy population(7175), and currently, there are no specific dietary guidelines available for individuals with ASD.

This gap in the existing guidelines contradicts the need for personalised and adapted management of this disorder, as emphasised by guidelines and action plans developed at the global(76), European(7780) and Italian(81,82) levels. These guidelines are endorsed by the most authoritative organisations in the field, that is, Autism Speaks(83).

In fact, ASD is a sensitive and lifelong condition that impacts every aspect of life. Hence, there is a crucial need for specific nutritional guidelines to improve the quality of life of those individuals, focusing on their health and inclusion in the community(Reference Conti, Breda and Basilico84). Considering the role of Zn, the frequent co-presence of ZD risk factors and FS problems in individuals with ASD highlights the potential need for formulating specific dietary recommendations for this population. Furthermore, considering the frequent occurrence of multiple comorbidities in individuals with ASD (e.g. recurrent infection, systemic and neurological inflammation, diarrhoea, microbiota alterations, leaky gut and metabolic disorders), which can either cause or contribute to ZD, relevant attention should be paid on Zn requirement.

Therefore, it is advisable to early detect Zn blood levels and assess dietary intake in individuals with ASD, ideally during infancy at the time of ASD diagnosis, to evaluate the presence of ZD. A potential workflow for proper management would involve following the principles and the structure of the Nutrition Care Process (NCP) developed by the American Dietetic Association, which includes nutrition assessment, nutrition diagnosis, nutrition intervention, nutrition monitoring and evaluation (followed by periodic re-assessment)(85,86) .

In reference to the results provided by the assessment phase (interview and clinical data collection), the next step would involve analysing plasma Zn levels(Reference Berger, Shenkin and Schweinlin87). If a ZD condition is identified, that is, <60 μg/dl in healthy adults(Reference Maxfield, Shukla and Crane88), the most appropriate strategy to address the deficiency would be planned. In this regard, the primary approach to be implemented is to improve dietary intake of Zn. If, on the other hand, the ZD condition is severe and/or persistent, potential supplementation strategies should be considered. In this scenario, recent European Society for Clinical Nutrition and Metabolism (ESPEN) recommendations suggest 0·5–1 mg/kg/d of elemental zinc (Zn2+) given orally for 3–4 months(Reference Berger, Shenkin and Schweinlin87). Moreover, the ESPEN panel emphasises that organic compounds (such as zinc histidinate, zinc gluconate and zinc orotate) are comparatively better tolerated than inorganic zinc sulphate and zinc chloride(Reference Berger, Shenkin and Schweinlin87). A reasonable approach in this case would be to gradually increase the zinc dosage from supplementation in parallel with an adequate dietary zinc intake, with periodic monitoring of blood levels(Reference Berger, Shenkin and Schweinlin87) to tailor treatment to the subject’s response while avoiding potential side effects. In fact, at doses >50 mg/d, GI symptoms, such as nausea, abdominal discomfort and diarrhoea, commonly occur(Reference Maxfield, Shukla and Crane88).

In conclusion, future perspectives could concern the development of a comprehensive screening tool that considers all the factors to which individuals with ASD are generally exposed, which can increase the risk of developing ZD, along with the individual’s current symptomatology. This screening tool would take the form of a questionnaire that provides a score indicating the risk of developing ZD, tailored specifically for individuals with ASD.

Acknowledgements

The authors would like to express their gratitude to Dr Irene Briata for providing valuable comments and suggestions on the review. Dr Briata is affiliated with the Post Graduate Course in Food Science and Human Nutrition, Università Statale di Milano, 20122 Milan, Italy. Also, the authors acknowledge the contribution of Dana El Masri for the English revision. Dr El Masri is affiliated with Laboratory of Dietetics and Clinical Nutrition, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, Pavia, Italy.

Financial support

This narrative review was conducted as a part of the FOOD-AUT project, funded by Pellegrini S.p.A. The project aims to improve the health status of individuals with autism spectrum disorders through a double intervention targeting canteen catering and caregivers. The project has resulted in the development of specific menus for canteens and two different nutritional recommendation documents for canteens and caregivers, addressing the nutritional and sensitive needs of this vulnerable population.

Competing interests

The authors declare no conflicts of interest.

Authorship

Maria Vittoria Conti, Sara Santero, Alessia Luzzi; methodology: Maria Vittoria Conti, Sara Santero, Alessia Luzzi; writing—original draft preparation: Sara Santero; writing—review and editing: Maria Vittoria Conti, Sara Santero, Alessia Luzzi; visualisation: Hellas Cena; supervision: Hellas Cena. All authors have read and agreed to the published version of the manuscript.

References

Kambe, T, Tsuji, T, Hashimoto, A, et al. (2015) The physiological, biochemical, and molecular roles of zinc transporters in zinc homeostasis and metabolism. Physiol Rev 95, 749784.CrossRefGoogle ScholarPubMed
Chasapis, CT, Ntoupa, PA, Spiliopoulou, CA, et al. (2020) Recent aspects of the effects of zinc on human health. Arch Toxicol 94, 14431460.CrossRefGoogle ScholarPubMed
Skalny, AV, Aschner, M & Tinkov, AA (2021) Zinc. Adv Food Nutr Res 96, 251310.CrossRefGoogle ScholarPubMed
Wessels, I, Fischer, HJ & Rink, L (2021) Dietary and physiological effects of zinc on the immune system. Annu Rev Nutr 41, 133175.CrossRefGoogle ScholarPubMed
Livingstone, C (2015) Zinc: physiology, deficiency, and parenteral nutrition. Nutr Clin Pract 30, 371382.CrossRefGoogle ScholarPubMed
Valenzuela-Zamora, AF, Ramírez-Valenzuela, DG & Ramos-Jiménez, A (2022) Food selectivity and its implications associated with gastrointestinal disorders in children with autism spectrum disorders. Nutrients 14 (13), 2660.CrossRefGoogle ScholarPubMed
Gallardo-Carrasco, MC, Jiménez-Barbero, JA, Bravo-Pastor, MDM, et al. (2022) Serum vitamin D, folate and fatty acid levels in children with autism spectrum disorder: a systematic review and meta-analysis. J Autism Dev Disord 52, 47084721.CrossRefGoogle ScholarPubMed
Marí-Bauset, S, Zazpe, I, Mari-Sanchis, A, et al. (2014) Food selectivity in autism spectrum disorders: a systematic review. J Child Neurol 29, 15541561.CrossRefGoogle ScholarPubMed
Alsufiani, HM, Alkhanbashi, AS, Laswad, NAB, et al. (2022) Zinc deficiency and supplementation in autism spectrum disorder and Phelan-McDermid syndrome. J Neurosci Res 100, 970978.CrossRefGoogle ScholarPubMed
Kuschner, ES, Eisenberg, IW, Orionzi, B, et al. (2015) A preliminary study of self-reported food selectivity in adolescents and young adults with autism spectrum disorder. Res Autism Spectr Disord 15–16, 5359.CrossRefGoogle ScholarPubMed
Roohani, N, Hurrell, R, Kelishadi, R, et al. (2013) Zinc and its importance for human health: an integrative review. J Res Med Sci 18, 144157.Google ScholarPubMed
Italian Society of Human Nutrition (2014) LARN: livelli di Assunzione di Riferimento di Nutrienti ed energia. IV revision, 2nd ed. Milan, Italy: SICS Editore.Google Scholar
American Psychiatric Association (2013) Diagnostic and Statistical Manual of Mental Disorders, 5th ed. Arlington, TX, USA: Pan American Medical Editorial.Google Scholar
Sharma, SR, Gonda, X & Tarazi, FI (2018) Autism Spectrum Disorder: classification, diagnosis and therapy. Pharmacol Ther 190, 91104.CrossRefGoogle ScholarPubMed
Sauer, AK, Malijauskaite, S, Meleady, P, et al. (2021) Zinc is a key regulator of gastrointestinal development, microbiota composition and inflammation with relevance for autism spectrum disorders. Cell Mol Life Sci 79 (1), 46.CrossRefGoogle ScholarPubMed
Bjørklund, G, Meguid, NA, El-Bana, MA, et al. (2020) Oxidative stress in autism spectrum disorder. Mol Neurobiol 57, 23142332.CrossRefGoogle ScholarPubMed
Li, YJ, Xie, XN, Lei, X, et al. (2020) Global prevalence of obesity, overweight and underweight in children, adolescents and adults with autism spectrum disorder, attention-deficit hyperactivity disorder: a systematic review and meta-analysis. Obes Rev 21 (12), e13123.CrossRefGoogle ScholarPubMed
Wohlmacher, SH (2017) Characterizing food selectivity in children with autism. Honors Theses and Capstones. 365. https://scholars.unh.edu/honors/365.Google Scholar
Bandini, LG, Anderson, SE, Curtin, C, et al. (2010) Food selectivity in children with autism spectrum disorders and typically developing children. J Pediatr 157, 259264.CrossRefGoogle ScholarPubMed
Siniscalco, D, Schultz, S, Brigida, AL, et al. (2018) Inflammation and neuro-immune dysregulations in autism spectrum disorders. Pharmaceuticals (Basel) 11 (2), 56.CrossRefGoogle ScholarPubMed
Ohashi, W & Fukada, T (2019) Contribution of zinc and zinc transporters in the pathogenesis of inflammatory bowel diseases. J Immunol Res 2019, 8396878. doi: 10.1155/2019/8396878.CrossRefGoogle ScholarPubMed
Fowlie, G, Cohen, N & Ming, X (2018) The perturbance of microbiome and gut-brain axis in autism spectrum disorders. Int J Mol Sci 19 (8), 2251.CrossRefGoogle ScholarPubMed
Dhaliwal, KK, Orsso, CE, Richard, C, et al. (2019) Risk factors for unhealthy weight gain and obesity among children with autism spectrum disorder. Int J Mol Sci 20 (13), 3285.CrossRefGoogle ScholarPubMed
Al-Ayadhi, L, Zayed, N, Bhat, RS, et al. (2021) The use of biomarkers associated with leaky gut as a diagnostic tool for early intervention in Autism Spectrum Disorder: a systematic review. Gut Pathog 13, 116.CrossRefGoogle Scholar
Srikantha, P & Mohajeri, MH (2019) The possible role of the microbiota-gut-brain-axis in autism spectrum disorder. Int J Mol Sci 20 (9), 2115.CrossRefGoogle ScholarPubMed
Fattorusso, A, Di Genova, L, Dell’Isola, GB, et al. (2019) Autism spectrum disorders and the gut microbiota. Nutrients 11 (3), 521.CrossRefGoogle ScholarPubMed
Matta, SM, Hill-Yardin, EL & Crack, PJ (2019) The influence of neuroinflammation in autism spectrum disorder. Brain Behav Immun 79, 7590.CrossRefGoogle ScholarPubMed
Saurman, V, Margolis, KG & Luna, RA (2020) Autism spectrum disorder as a brain-gut-microbiome axis disorder. Dig Dis Sci 65, 818828.CrossRefGoogle ScholarPubMed
Leader, G, Barrett, A, Ferrari, C, et al. (2021) Quality of life, gastrointestinal symptoms, sleep problems, social support, and social functioning in adults with autism spectrum disorder. Res Dev Disabil 112, 103915.CrossRefGoogle ScholarPubMed
Bhandari, R, Paliwal, JK & Kuhad, A (2020) Neuropsychopathology of autism spectrum disorder: complex interplay of genetic, epigenetic, and environmental factors. Adv Neurobiol 24, 97141.CrossRefGoogle ScholarPubMed
Zheng, Z, Zhu, T, Qu, Y, et al. (2016) Blood glutamate levels in autism spectrum disorder: a systematic review and meta-analysis. PLoS One 11 (7), e0158688.CrossRefGoogle ScholarPubMed
Cortese, S, Gabellone, A, Marzulli, L, et al. (2022) Association between autism spectrum disorder and diabetes: systematic review and meta-analysis. Neurosci Biobehav Rev 136, 104592.CrossRefGoogle ScholarPubMed
Vela, G, Stark, P, Socha, M, et al. (2015) Zinc in gut–brain interaction in autism and neurological disorders. Neural Plast 2015.CrossRefGoogle ScholarPubMed
Olechnowicz, J, Tinkov, A, Skalny, A, et al. (2018) Zinc status is associated with inflammation, oxidative stress, lipid, and glucose metabolism. J Physiol Sci 68, 1931.CrossRefGoogle ScholarPubMed
Tamura, Y (2021) The role of zinc homeostasis in the prevention of diabetes mellitus and cardiovascular diseases. J Atheroscler Thromb 28, 11091122.CrossRefGoogle ScholarPubMed
Manco, M, Guerrera, S, Ravà, L, et al. (2021) Cross-sectional investigation of insulin resistance in youths with autism spectrum disorder. Any role for reduced brain glucose metabolism? Transl Psychiatry 11 (1), 229.CrossRefGoogle ScholarPubMed
Berezin, AE, Berezin, AA & Lichtenauer, M (2020) Emerging role of adipocyte dysfunction in inducing heart failure among obese patients with prediabetes and known diabetes mellitus. Front Cardiovasc Med 7, 583175.CrossRefGoogle ScholarPubMed
Sanctuary, MR, Kain, JN, Angkustsiri, K, et al. (2018) Dietary considerations in autism spectrum disorders: the potential role of protein digestion and microbial putrefaction in the gut-brain axis. Front Nutr 5, 40.CrossRefGoogle ScholarPubMed
Gillberg, C, Fernell, E, Kočovská, E, et al. (2017) The role of cholesterol metabolism and various steroid abnormalities in autism spectrum disorders: a hypothesis paper. Autism Res 10, 10221044.CrossRefGoogle ScholarPubMed
Luçardo, JDC, Monk, GF, Dias, MDS, et al. (2021) Interest in food and triglyceride concentrations in children and adolescents with autistic spectrum disorder. J Pediatr 97, 103108.CrossRefGoogle ScholarPubMed
Shi, Y, Zou, Y, Shen, Z, et al. (2020) Trace elements, PPARs, and metabolic syndrome. Int J Mol Sci 21 (7), 2612.CrossRefGoogle ScholarPubMed
Santos, HO, Teixeira, FJ, Schoenfeld, BJ, et al. (2020) Dietary vs. pharmacological doses of zinc: a clinical review. Clin Nutr 39, 13451353.CrossRefGoogle ScholarPubMed
Bishop, L, Charlton, RA, McLean, KJ, et al. (2023) Cardiovascular disease risk factors in autistic adults: the impact of sleep quality and antipsychotic medication use. Autism Res 16, 569579.CrossRefGoogle ScholarPubMed
Chu, A, Foster, M & Samman, S (2016) Zinc status and risk of cardiovascular diseases and type 2 diabetes mellitus – a systematic review of prospective cohort studies. Nutrients 8 (11), 707.CrossRefGoogle ScholarPubMed
Manna, P & Jain, SK (2015) Obesity, oxidative stress, adipose tissue dysfunction, and the associated health risks: causes and therapeutic strategies. Metab Syndr Relat Disord 13, 423444.CrossRefGoogle ScholarPubMed
Indika, NLR, Frye, RE, Rossignol, DA, et al. (2023) The rationale for vitamin, mineral, and cofactor treatment in the precision medical care of autism spectrum disorder. J Pers Med 13 (2), 252.CrossRefGoogle ScholarPubMed
Kumbargere Nagraj, S, George, RP, Shetty, N, et al. (2017) Interventions for managing taste disturbances. Cochrane Database Syst Rev 12, 12.Google ScholarPubMed
Nishiguchi, Y, Ohmoto, M, Koki, J, et al. (2016) Bcl11b/Ctip2 is required for development of lingual papillae in mice. Dev Biol 416, 98110.CrossRefGoogle ScholarPubMed
Ikeda, A, Sekine, H, Takao, K, et al. (2013) Expression and localization of taste receptor genes in the vallate papillae of rats: effect of zinc deficiency. Acta Otolaryngol 133, 957964.CrossRefGoogle ScholarPubMed
Onoda, K, Hirai, R, Takao, K, et al. (2011) Patients with hypogeusia show changes in expression of T2R taste receptor genes in their tongues. Laryngoscope 121, 25922597.CrossRefGoogle ScholarPubMed
Fábián, TK, Beck, A, Fejérdy, P, et al. (2015) Molecular mechanisms of taste recognition: considerations about the role of saliva. Int J Mol Sci 16, 59455974.CrossRefGoogle ScholarPubMed
Wang, Y, Zajac, AL, Lei, W, et al. (2019) Metal ions activate the human taste receptor TAS2R7. Chem Senses 44, 339347.CrossRefGoogle ScholarPubMed
Braud, A & Boucher, Y (2020) Taste disorder’s management: a systematic review. Clin Oral Investig 24, 18891908.CrossRefGoogle ScholarPubMed
Santos, HO (2022) Therapeutic supplementation with zinc in the management of COVID-19–related diarrhea and ageusia/dysgeusia: mechanisms and clues for a personalized dosage regimen. Nutr Rev 80, 10861093.CrossRefGoogle ScholarPubMed
Baltaci, AK, Mogulkoc, R & Baltaci, SB (2019) Review: the role of zinc in the endocrine system. Pak J Pharm Sci 32, 231239.Google ScholarPubMed
Levenson, CW (2003) Zinc regulation of food intake: new insights on the role of neuropeptide Y. Nutr Rev 61, 247249.Google ScholarPubMed
Komai, M, Goto, T, Ohinata, K, et al. (2018) Clarification of the mechanism involved in orexigenic action by oral zinc ingestion. Yakugaku Zasshi 138, 10111016. Japanese.CrossRefGoogle ScholarPubMed
Sarcia, B (2021) The impact of applied behavior analysis to address mealtime behaviors of concern among individuals with autism spectrum disorder. Psychiatr Clin North Am 44, 8393.CrossRefGoogle ScholarPubMed
Kodak, T & Piazza, CC (2008) Assessment and behavioral treatment of feeding and sleeping disorders in children with autism spectrum disorders. Child Adolesc Psychiatric Clin N Am 17, 887905.CrossRefGoogle Scholar
Baj, J, Flieger, W, Flieger, M, et al. (2021) Autism spectrum disorder: trace elements imbalances and the pathogenesis and severity of autistic symptoms. Neurosci Biobehav Rev 129, 117132.CrossRefGoogle ScholarPubMed
Maares, M & Haase, H (2020) A guide to human zinc absorption: general overview and recent advances of in vitro intestinal models. Nutrients 12 (3), 762.CrossRefGoogle ScholarPubMed
Shkembi, B & Huppertz, T (2021) Influence of dairy products on bioavailability of zinc from other food products: a review of complementarity at a meal level. Nutrients 13 (12), 4253.CrossRefGoogle Scholar
Bel-Serrat, S, Stammers, AL, Warthon-Medina, M, et al. (2014) Factors that affect zinc bioavailability and losses in adult and elderly populations. Nutr Rev 72, 334352.CrossRefGoogle ScholarPubMed
Agnoli, C, Baroni, L, Bertini, I, et al. (2017) Position paper on vegetarian diets from the working group of the Italian Society of Human Nutrition. Nutr Metab Cardiovasc Dis 27, 10371052.CrossRefGoogle Scholar
Melse-Boonstra, A (2020) Bioavailability of micronutrients from nutrient-dense whole foods: zooming in on dairy, vegetables, and fruits. Front Nutr 7, 101.CrossRefGoogle Scholar
Platel, K & Srinivasan, K (2016) Bioavailability of micronutrients from plant foods: an update. Crit Rev Food Sci Nutr 56, 16081619.CrossRefGoogle ScholarPubMed
Centro Ricerca Alimenti e Nutrizione (2018) LINEE GUIDA PER UNA SANA ALIMENTAZIONE. Revision 2018. Available at: https://www.crea.gov.it/web/alimenti-e-nutrizione/-/linee-guida-per-una-sana-alimentazione-2018 Google Scholar
Park, HJ, Choi, SJ, Kim, Y, et al. (2020) Mealtime behaviors and food preferences of students with autism spectrum disorder. Foods 10 (1), 49.CrossRefGoogle ScholarPubMed
Ahumada, D, Guzmán, B, Rebolledo, S, et al. (2022) Eating patterns in children with autism spectrum disorder. Healthcare (Basel) 10, 1829.CrossRefGoogle ScholarPubMed
Jones, RA, Downing, K, Rinehart, NJ, et al. (2017) Physical activity, sedentary behavior and their correlates in children with autism spectrum disorder: a systematic review. PLoS One 12 (2), e0172482.CrossRefGoogle ScholarPubMed
World Health Organization/Food and Agricultural Organization (2004) Vitamin and Mineral Requirements in Human Nutrition, 2nd ed. Geneva, Switzerland: World Health Organization.Google Scholar
Institute of Medicine (2002) Dietary Reference Intakes of Vitamin A, Vitamin K, Arsenic Boron, Chromium, Copper, Iodine, Iron, Manganese, Molybdenum, Nickel, Silicon, Vanadium, and Zinc. Washington, DC: National Academy Press.Google Scholar
International Zinc Nutrition Consultative Group (IZiNCG), Brown, KH, Rivera, JA et al. (2004) International Zinc Nutrition Consultative Group (IZiNCG) technical document #1. Assessment of the risk of zinc deficiency in populations and options for its control. Food Nutr Bull 25 (Suppl 2), S99S203.Google Scholar
European Food Safety Authority (EFSA) Panel on Dietetic Products, Nutrition and Allergies (NDA) (2014) Scientific opinion on Dietary Reference values for zinc. EFSA J 12, 176.Google Scholar
LARN MINERALI (2019) Assunzione raccomandata per la popolazione (PRI) e assunzione adeguata (AI). Available at: https://sinu.it/2019/07/09/minerali-assunzione-raccomandata-per-la-popolazione-pri-e-assunzione-adeguataai/ Google Scholar
Report by the Secretariat (2013) Comprehensive and coordinated efforts for the management of Autism Spectrum Disorders, WHO Executive Board, 133rd session, Provisional agenda item 6.1,8 April 2013. Available at: https://apps.who.int/gb/ebwha/pdf_files/eb133/b133_4-en.pdf Google Scholar
WHO European framework for action on mental health 2021–2025 (2021) Draft for the Seventy-first Regional Committee for Europe. Copenhagen: WHO Regional Office for Europe. Licence: CC BY-NC-SA 3.0 IGOGoogle Scholar
Fuentes, J, Hervás, A, Howlin, P, et al. (2021) ESCAP practice guidance for autism: a summary of evidence-based recommendations for diagnosis and treatment. Eur Child Adolesc Psychiatry 30, 961984.CrossRefGoogle ScholarPubMed
European Pact for Mental Health and Well-being (EU High-level Conference, Brussels, 12–13 June 2008). Available at: https://ec.europa.eu/health/ph_determinants/life_style/mental/docs/pact_en.pdf Google Scholar
European Framework for Mental Health and Well-Being (EU Joint Action on Mental Health and Wellbeing, Final Conference, Brussels, 21–22 January 2016). Available at: https://ec.europa.eu/research/participants/data/ref/h2020/other/guides_for_applicants/h2020-SC1-BHC-22-2019-framework-for-action_en.pdf Google Scholar
Conferenza Unificata 10 Maggio 2018. Aggiornamento delle linee di indirizzo per la promozione ed il miglioramento della qualità e dell’appropriatezza degli interventi assistenziali nei Disturbi dello Spettro Autistico. Available at: file:///C:/Users/Santero/Downloads/News%202018-05-14%2053CU_100518%20(1).pdf Google Scholar
Piano di azioni nazionale per la salute mentale. Available at: https://www.salute.gov.it/imgs/C_17_pubblicazioni_1905_allegato.pdf Google Scholar
Autism Speaks website. Available at: https://www.autismspeaks.org/ Google Scholar
Conti, MV, Breda, C, Basilico, S, et al. (2023) Dietary recommendations to customize canteen menus according to the nutritional and sensory needs of individuals with Autism Spectrum Disorder. EWD. Manuscript accepted, in press.CrossRefGoogle Scholar
Writing Group of the Nutrition Care Process/Standardized Language Committee (2008) Nutrition care process and model part I: the 2008 update. J Am Diet Assoc 108, 11131117.CrossRefGoogle Scholar
Writing Group of the Nutrition Care Process/Standardized Language Committee (2008) Nutrition care process part II: using the International Dietetics and Nutrition Terminology to document the nutrition care process. J Am Diet Assoc 108, 12871293.CrossRefGoogle Scholar
Berger, MM, Shenkin, A, Schweinlin, A, et al. (2022) ESPEN micronutrient guideline. Clin Nutr 41, 13571424.CrossRefGoogle ScholarPubMed
Maxfield, L, Shukla, S & Crane, JS (2022) Zinc deficiency. In: StatPearls [Internet]. Treasure Island, FL: StatPearls Publishing.Google Scholar
Figure 0

Table 1. The multiple roles played by Zn in taste perception

Figure 1

Table 2. Factors with a positive, negative or ‘neutral’ effect on dietary Zn bioavailability