Hostname: page-component-cb9f654ff-kl2l2 Total loading time: 0 Render date: 2025-08-08T08:57:17.765Z Has data issue: false hasContentIssue false

Zinc ion dyshomeostasis in autism spectrum disorder

Published online by Cambridge University Press:  13 March 2025

Asma Ahmadani
Affiliation:
Department of Nutrition and Health, College of Medicine & Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
Monia Kittana
Affiliation:
Department of Nutrition and Health, College of Medicine & Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
Farah Al-Marzooq
Affiliation:
Department of Microbiology and Immunology, College of Medicine & Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
Sandeep B. Subramanya
Affiliation:
Department of Physiology, College of Medicine & Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
Maria Cristina D’Adamo
Affiliation:
Department of Medicine & Surgery, LUM University “GIUSEPPE DEGENNARO”, Casamassima, BA, Italy
Amita Attlee*
Affiliation:
Department of Nutrition and Health, College of Medicine & Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
Mauro Pessia*
Affiliation:
Department of Physiology, College of Medicine & Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
*
Corresponding authors: Mauro Pessia; Email: mauro@uaeu.ac.ae, Amita Attlee; Email: amita.attlee@uaeu.ac.ae
Corresponding authors: Mauro Pessia; Email: mauro@uaeu.ac.ae, Amita Attlee; Email: amita.attlee@uaeu.ac.ae
Rights & Permissions [Opens in a new window]

Abstract

Autism spectrum disorder (ASD) is a neurodevelopmental disorder with significant social, communicative, and behavioral challenges, and its prevalence is increasing globally at an alarming rate. Children with ASD often have nutritional imbalances, and multiple micronutrient deficiencies. Among these, zinc (Zn2+) deficiency is prominent and has gained extensive scientific interest over the past few years. Zn2+ supports numerous proteins, including enzymes and transcription factors, and controls neurogenesis and cell differentiation. It modulates synaptic transmission and plasticity by binding to receptors, ion channels, and transporters. These interactions are crucial, as changes in these processes may contribute to cognitive and behavioral abnormalities in neurodevelopmental disorders, including ASD. Notably, mutations in genes linked to ASD result in Zn2+ dyshomeostasis, altering pivotal biological processes. In addition, Zn2+ promotes gut health by maintaining gut wall integrity, preventing inflammation and leaky gut, preventing translocation of gut bacteria and their metabolites into systemic circulation, and supporting cognitive processes via the gut–brain axis. Zn2+ deficiency during pregnancy alters gut microbiota composition, induces pro-inflammatory cytokine production, may affect neuronal functioning, and is associated with ASD etiology in offspring, as well as the exacerbation of autistic traits in genetically predisposed children. This review focuses on Zn2+ dyshomeostasis, discussing various Zn2+-dependent dysfunctions underlying distinct autistic phenotypes and describing recent progress in the neurobiology of individuals with ASD and animal models.

Information

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 (https://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), 2025. Published by Cambridge University Press on behalf of The Nutrition Society

Introduction

Autism spectrum disorder (ASD) comprises a group of neurodevelopmental disorders characterized by highly variable clinical manifestations, including deficits in social interactions, interpersonal communication, and repetitive and stereotypical behaviors(1,Reference Mubashir, Farrugia, Coretti, Pessia and D’Adamo2) . Since no unique biomarkers have been established, diagnosis is primarily based on the assessment of behavioral phenotypes(Reference Lord, Elsabbagh, Baird and Veenstra-Vanderweele3).

The global prevalence of ASD is rapidly increasing(Reference Fombonne4). It is a multifactorial disorder, and owing to its complex heterogeneity, its exact etiology is not well established(Reference Grabrucker5). However, genetic, epigenetic, environmental, and nutritional factors have been implicated in its etiology(Reference Bhandari, Paliwal, Kuhad, Essa and Qoronfleh6Reference Yoon, Choi, Lee and Do11). More recently, zinc (Zn2+) dyshomeostasis has been proposed as a significant risk factor for ASD development(Reference Alsufiani, Alkhanbashi, Laswad, Bakhadher, Alghamdi and Tayeb12,Reference Babaknejad, Sayehmiri, Sayehmiri, Mohamadkhani and Bahrami13) . Notably, Zn2+ is a trace mineral essential for growth and development(Reference Maret and Sandstead14) and is increasingly acknowledged as a pivotal signaling molecule(Reference Hara, Takeda, Takagishi, Fukue, Kambe and Fukada15Reference Fukada, Yamasaki, Nishida, Murakami and Hirano17).

The United Nations Food and Agriculture Organization has indicated that the Zn2+ content in national food supplies might be inadequate to meet the requirements of approximately 15–20% of the world’s population(Reference Wessells and Brown18). The World Health Organization estimates that approximately 30% of the world’s population is Zn2+-deficient, with the prevalence ranging between 4% and 73% depending on the region(Reference Kumera, Awoke, Melese, Eshetie, Mekuria and Mekonnen19,Reference Prasad and Collins20) . A recent Irish study revealed that 58% of women face the risk of insufficient Zn2+ intake, with 29% potentially being Zn2+ deficient. Notably, pregnant women exhibited lower prevalence rates, which is attributable to a more frequent supplement consumption(Reference De Benedictis, Trame, Rink and Grabrucker21). In addition, people living in South and Southeast Asia, sub-Saharan Africa, and Central America were identified as being at the highest risk of inadequate Zn2+ intake on the basis of its availability in national food supplies(Reference Wessells and Brown18). Zn2+ deficiency can arise due to several factors, including decreased intake (malnutrition, anorexia, parenteral nutrition), decreased bioavailability (vegetarian diets or the consumption of Zn2+ uptake inhibitors, such as iron, copper (Cu2+), and calcium), decreased absorption (inflammatory bowel disease, chronic pancreatic disease, celiac sprue, alcoholism), increased loss (renal disorders, burns, exudative skin disease, chronic blood loss or gastro-intestinal loss, alcoholics with and without liver disease), or increased requirements (pregnancy, lactation, neoplasia, chronic infections, or inflammation)(Reference Scarpellini, Balsiger, Maurizi, Rinninella, Gasbarrini and Giostra22). Furthermore, Zn2+ deficiencies resulting in acrodermatitis enteropathica may arise from genetic mutations in SLC39A4, which encodes the Zn2+ importer ZIP4, a transmembrane protein that mediates the transport of multiple ions including Zn2+(Reference Zhong, Yang, Zhu, Huang and Chen23,Reference McClain, Vatsalya and Cave24) . The clinical manifestations of Zn2+ deficiency in pregnant and pediatric populations include delayed or abnormal fetal growth, low fetal cognition, low birth weight, preterm labor, growth retardation, hypogonadism, dry skin, acrodermatitis enteropathica, delayed wound healing, diarrhea, and increased allergic and inflammatory reactions(Reference Corbo and Lam25Reference Willoughby and Bowen28). Conversely, in adults, Zn2+ is essential for mental well-being; consequently, its deficiency may be associated with attention deficits, altered behavioral and emotional responses, and depression(Reference Grønli, Kvamme, Friborg and Wynn29,30) . In animal models, Zn2+ deficiency induced by a Zn2+-depleted diet facilitated the development of depressive symptoms, deficits in learning, memory and attention, aggressive and anxious behaviors, in addition to causing wasting and stunting(Reference Hagmeyer, Haderspeck and Grabrucker31). These behaviors seem to be exacerbated in ASD and may arise from restrictive/inadequate food intake(Reference Sweetman, O’Donnell, Lalor, Grant and Greaney32) and eating/feeding problems(Reference Cherif, Boudabous, Khemekhem, Mkawer, Ayadi and Moalla33,Reference Sharp, Berry, McCracken, Nuhu, Marvel and Saulnier34) in addition to other underlying conditions. Individuals with ASD are vulnerable to oxidative stress, further increasing their body’s demand for Zn2+ and worsening its deficiency(Reference Bjørklund, Meguid, El-Bana, Tinkov, Saad and Dadar35). Owing to its ubiquitous presence and crucial involvement in varied biological processes, Zn2+ is vital for growth and development(Reference Maret and Sandstead14). It is involved in a multitude of catalytic, structural, and functional regulations, including DNA metabolism, protein synthesis, immunity regulation, reproduction, vision, taste, neuronal growth, and neurotransmission(Reference Maret and Sandstead14). In addition, it is well known for its role in maintaining intestinal health and microbial homeostasis(Reference Scarpellini, Balsiger, Maurizi, Rinninella, Gasbarrini and Giostra22,Reference Vela, Stark, Socha, Sauer, Hagmeyer and Grabrucker36) .

A comprehensive understanding of crucial physiological and pathological processes regulated by Zn2+ is a pivotal step in elucidating the distinct neurobiological mechanisms of ASD, establishing preventive strategies, and identifying original therapeutic solutions for this disorder. Therefore, this paper aimed to concisely discuss clinical findings related to ASD and elaborate on the most relevant physiological roles of Zn2+, while thoroughly reviewing recent progress in the neurobiology of ASD in which Zn2+-dependent pathways have been implicated. Nutritional aspects and interactions between gut microbiota and the brain are particularly emphasized.

Methodology

This narrative review began with a literature search in July 2021, following a non-systematic approach. Titles and abstracts were searched across PubMed, Cochrane Library, and Google Scholar, covering publications up to June 2024. The review included original research, systematic reviews, meta-analyses, narrative reviews, study protocols, pilot studies, and randomized controlled trials (RCT). The search utilized keywords such as but not limited to: “autism spectrum disorder (ASD)” AND “prevalence” OR “zinc deficiency” OR “zinc-dyshomeostasis” OR “neurodevelopmental disorders” OR “gut–brain axis” OR “gut microbiota” OR “microbial dysbiosis” OR “SHANK proteins” OR “neurophysiology” OR “nutritional imbalance” OR “prenatal zinc deficiency” OR “zinc supplementation” OR “gut inflammation” OR “leaky gut syndrome” OR “tight junction complexes” OR “zinc transporters” OR “environmental factors in ASD” OR “genetic factors in ASD” OR “zinc and brain function” OR “zinc in child development” OR “therapeutic zinc use” OR “rodent models of ASD” OR “gut integrity” OR “systemic inflammation in ASD” OR “autism pathophysiology.”

Articles were initially screened by title and abstract, excluding those misaligned with the manuscript’s themes. The remaining articles underwent full-text review. The inclusion criteria were: (1) articles addressing autism, its clinical characteristics, and prevalence; (2) studies on clinical findings related to ASD, zinc’s physiological roles, the neurobiology involving zinc-dependent pathways, and zinc’s impact on gut physiology and microbiota; (3) full-text articles; and (4) studies published in English. Overall, a total of 226 articles were selected.

Ultimately, studies were included on the basis of their alignment with the research objectives, methodological rigor, and overall contribution to the understanding of zinc dyshomeostasis in ASD, as shown in the PRISMA diagram (Fig. 1).

Figure 1. PRISMA flow diagram illustrating the search process, detailing the identification, screening, and quality assessment phases, which ultimately resulted in the inclusion of 224 studies and two reports in the final review.

ASD—current status of knowledge

Prevalence: In 2023, a three-level meta-analysis estimated the global prevalence of ASD to be approximately seventy-two cases per 10,000 individuals (95% confidence interval [CI]: 61–85)(Reference Talantseva, Romanova, Shurdova, Dolgorukova, Sologub and Titova37). The global prevalence of ASD varies significantly among different regions and countries. Although the Global North employs novel techniques, such as deep learning models, early biomarker identification, and integration of multimodal data to facilitate timely ASD diagnosis, certain challenges persist in other countries. These include disparities in diagnostic criteria, public awareness, healthcare infrastructure, and lengthy waiting periods, which collectively contribute to the diverse prevalence rates observed across the globe(Reference Fombonne, MacFarlane and Salem38).

In North America, the USA reported that approximately one in thirty-six children aged 8 years were diagnosed with ASD by eleven monitoring sites across the USA in 2020(Reference Maenner39). According to the Canadian Health Survey on Children and Youth reports, Canada also shows a notable prevalence, with an estimated one in fifty (2%) Canadian children aged 1–17 years diagnosed with ASD in 2019(40).

A district-based study in Sweden reported the highest ASD prevalence of 3·66% among preschoolers(Reference Linnsand, Gillberg, Nilses, Hagberg and Nygren41). The United Kingdom (UK) has one of the highest countrywide prevalence rates. According to recent research, approximately 2·94% (one in thirty-four) of 10–14 year olds were diagnosed with ASD in 2018 in the UK(Reference O’Nions, Petersen, Buckman, Charlton, Cooper and Corbett42). Other European countries, such as Sweden and Ireland, also report high prevalence rates of 661·85 and 583·69 per 100,000 people, 1 in 151 and 1 in 171, respectively(Reference Zeidan, Fombonne, Scorah, Ibrahim, Durkin and Saxena43).

Asia also has varying prevalence rates. A region-based study in Japan reported a prevalence of 3·22% in the total 5-year-old population screened for ASD between 2013 and 2016, with a cumulative incidence of 1·31%(Reference Saito, Hirota, Sakamoto, Adachi, Takahashi and Osato-Kaneda44). While South Korea had an estimated incidence of 9·4/1000 (1 in 106) by 2020, Singapore’s prevalence rate was estimated to be comparatively higher at 3·2% of children (0–6 years) diagnosed with ASD in 2020(Reference Sung, Magiati, Goh, Fung, Aljunied and Phua45).

Among South Asian countries, India had an estimated prevalence of 1 in 125 children aged 2–6 years in 2021(Reference Tiwari, Purkayastha and Gulati46), while in Bangladesh and Nepal, the prevalence in 2018 was 0·075% and 0·3%, respectively(Reference Rasool, Rasool, Siddique, Bajwa, Khan and Rasool47). The lower rates in the latter two countries might reflect underdiagnosis rather than an actual lower incidence. In Oceania, Australia had a high prevalence of 4·36% in the older child population (aged 12–13 years) compared with 2·6% among those diagnosed under 5 years of age(Reference May, Brignell and Williams48). New Zealand reported a lower percentage of about 1% or 1 of 102 cases in the total population of 8 year olds(Reference Bowden, Thabrew, Kokaua, Audas, Milne and Smiler49). African countries generally reported lower prevalence rates, which may be due to limited awareness and restricted access to healthcare and diagnostic services. Nigeria, for instance, has a nationwide prevalence of approximately 2·3%(Reference Lagunju, Bella-Awusah and Omigbodun50), while South Africa’s prevalence in the Western Cape Province was as low as 0·08%(Reference Pillay, Duncan and de Vries51). Systematic reviews from Gulf countries revealed that 1 in 146 children had ASD in the United Arab Emirates (UAE)(Reference Virolainen, Hussien and Dalibalta52). Qatar was reported to have the highest prevalence at 1·14% (0·89–1·46%)(Reference Alshaban, Aldosari, Al-Shammari, El-Hag, Ghazal and Tolefat53) followed by Saudi Arabia (2·81 in 1000)(Reference Sabbagh, Al-Jabri, Alsulami, Hashem, Aljubour and Alamoudi54), Oman (20·3 in 10,000), and Bahrain (4·3:10,000)(Reference Salhia, Al-Nasser, Taher, Al-Khathaami and El-Metwally55,Reference Al-Mamri, Idris, Dakak, Al-Shekaili, Al-Harthi and Alnaamani56) . Generally, boys have an approximately four times greater chance of being diagnosed with ASD than girls(Reference Alsufiani, Alkhanbashi, Laswad, Bakhadher, Alghamdi and Tayeb12), with an approximate male-to-female ratio of 4·3:1 globally(Reference Talantseva, Romanova, Shurdova, Dolgorukova, Sologub and Titova37). Dissimilarities in synaptic and cerebral plasticity between the sexes could account for such ratio differences(Reference Mottron, Duret, Mueller, Moore, Forgeot d’Arc and Jacquemont57).

Genetics: Mutations in genes encoding proteins responsible for the normal functioning of the neuronal pathways and synapses in the central nervous system (CNS) are associated with ASD(Reference Daghsni, Rima, Fajloun, Ronjat, Brusés and M’rad58,Reference Zoghbi and Bear59) . However, individuals with ASD exhibit significant heterogeneity in their genetic makeup and phenotypic expression, making ASD a disorder with a complex multifactorial etiology(Reference Lyall, Croen, Daniels, Fallin, Ladd-Acosta and Lee60).

Furthermore, as ASD prevalence is higher among consanguineous families and it aggregates within families, it is essential to understand its heritability(Reference Sandin, Lichtenstein, Kuja-Halkola, Hultman, Larsson and Reichenberg61,Reference Yousuf, Asif, Husain and Rafiq62) . Several genetic mutations have been identified in affected individuals(Reference Yu, Chahrour, Coulter, Jiralerspong, Okamura-Ikeda and Ataman63Reference Muhle, Trentacoste and Rapin66). Notably, the function of some of these gene products is modulated by Zn2+, such as the SH3 and multiple ankyrin repeat domain proteins (SHANK3) and the γ-aminobutyric acid receptor (GABAA). Mutations in genes encoding proteins that form ion channels have also been associated with ASD and the activity of several of these channel types is modulated by Zn2+(Reference Ambrosini, Sicca, Brignone, D’Adamo, Napolitano and Servettini67Reference D’Adamo, Moro, Imbrici, Martino, Roscini and Santorelli75). The identified candidate genes are currently being used to bolster the findings pertaining to the epigenetics of ASD(Reference Rylaarsdam and Guemez-Gamboa76). Noteworthily, a comprehensive genetic database is offered by the Simons Foundation Autism Research Initiative (SFARI)(77). This database is an invaluable resource for autism research, offering extensive genetic, phenotypic, and neuropsychological datasets to uncover the genetic and molecular underpinnings of ASD (e.g., Simons Simplex Collection, SFARI Variant Repository, and SPARK). By enabling access to large-scale, high-quality data, and tools such as induced pluripotent stem cells, SFARI facilitates hypothesis-driven, collaborative, and translational research advancing personalized medicine approaches and fostering scientific discovery in ASD.

Environment: Although several studies claim that genetics and epigenetics are the leading causes of ASD, an equal number of conflicting studies hold environmental factors accountable as players in ASD etiology(Reference Lyall, Croen, Daniels, Fallin, Ladd-Acosta and Lee60). Among these, viral infections, brain inflammation, defects in the placental or blood–brain barrier, environmental toxins, infections during pregnancy, birth status, over-immunization, age, and education are implicated(Reference Bölte, Girdler and Marschik78,Reference Ratajczak79) . Maternal educational levels in Jordan revealed that illiterate mothers and mothers with primary/secondary education were twelve times and three times more likely to have a child with autism, respectively, compared with mothers with a university education [OR: 12·25 (95% CI: 1·18–126·91) and OR: 3·319 (95% CI: 0·34–40·62)](Reference Alkhalidy, Abushaikha, Alnaser, Obeidat and Al-Shami80). Children born through vaginal delivery were less likely to be autistic compared with those born through caesarean delivery (OR: 0·273 [95% CI: 0·105–0·712)](Reference Alkhalidy, Abushaikha, Alnaser, Obeidat and Al-Shami80). In addition to several other reported causes, advanced maternal (> 40 years) and paternal (> 50 years) ages are believed to be independent risk factors for ASD(Reference Lord, Elsabbagh, Baird and Veenstra-Vanderweele3).

Nutrition: Among the various maternal dietary patterns during pregnancy, both inadequate nutritional intake(Reference Cortés-Albornoz, García-Guáqueta, Velez-van-Meerbeke and Talero-Gutiérrez81) and consumption of a Western diet have been positively associated with the etiology of neurological disorders, particularly ASD(Reference Vecchione, Wang, Rando, Chavarro, Croen and Fallin82,Reference Veniaminova, Cespuglio, Cheung, Umriukhin, Markova and Shevtsova83) . Nutritional deficiencies in folic acid, iron, vitamin D, and Zn2+ during pregnancy increased the risk of neurodevelopmental disorders in the fetus(Reference Gogou and Kolios84); whereas, multivitamin use during pregnancy, especially folic acid, and omega-3, played a protective role(Reference Gogou and Kolios84,Reference Bragg, Chavarro, Hamra, Hart, Tabb and Weisskopf85) . Zn2+ affects fertility and is particularly important during gestation and for fetal growth and development(Reference Akdas and Yazihan86). In later stages of pregnancy, Zn2+ deficiency has been linked to impaired neuronal growth, brain function, and synaptogenesis, potentially leading to impaired postnatal brain function and behavioral abnormalities(Reference Grzeszczak, Kwiatkowski and Kosik-Bogacka87). Overall, Zn2+ deficiency can cause fetal malformations, growth retardation, pregnancy or birth complications, and in severe cases, it can lead to embryonic, fetal, or maternal deaths(Reference Akdas and Yazihan86,Reference Iqbal and Ali88) . Studies have shown that low maternal cord Zn2+ levels are associated with adverse pregnancy outcomes(Reference Akdas and Yazihan86,Reference Iqbal and Ali88) . Furthermore, abnormal Zn2+ levels in the plasma and brain have been consistently observed in children with autism spectrum disorder (ASD-C)(Reference Babaknejad, Sayehmiri, Sayehmiri, Mohamadkhani and Bahrami13), possibly reflecting a broader underlying imbalance. Since Zn2+ dyshomeostasis is prevalent in individuals with ASD and has been frequently reported in the literature, this review focused on the role of this essential trace mineral in ASD.

Zinc levels in ASD

The Zn2+ status in an individual can be assessed using body fluids, such as urine, plasma, serum, red blood cells, whole blood, or hair(Reference Saghazadeh, Ahangari, Hendi, Saleh and Rezaei89). The levels of Zn2+ assessed in these samples are highly variable among ASD-C(Reference Alsufiani, Alkhanbashi, Laswad, Bakhadher, Alghamdi and Tayeb12,Reference Babaknejad, Sayehmiri, Sayehmiri, Mohamadkhani and Bahrami13,Reference Faber, Zinn, Kern and Skip Kingston90Reference Crăciun, Bjørklund, Tinkov, Urbina, Skalny and Rad93) . In a systematic review comparing Zn2+ levels between individuals with ASD and typically developing (TD) individuals, 36% of the studies reported significantly lower levels of Zn2+, with the remaining studies indicating a lower presence of Zn2+(Reference do Nascimento, Oliveira Silva, de Morais and de Rezende94). Two case-control studies from China found significantly lower blood Zn2+ levels among ASD-C than among TD children(Reference Guo, Li, Zhang, Chen, Dai and Liu95). In addition, associations between decreased serum Zn2+ concentrations (≤ 1.28 × 10−5 mol/L) and an increased risk and severity of ASD, indicating a potential role of Zn2+ in the pathophysiology of ASD(Reference Wu, Wang, Yan, Jia, Zhang and Han96) were identified. A meta-analysis of twenty-six studies further confirmed significantly lower blood levels of Zn2+ in individuals with ASD (n = 513) than in controls (n = 333), with a mean difference of −0·361 (95% CI: −0·668 to −0·055) in 50% of the investigations(Reference Saghazadeh, Ahangari, Hendi, Saleh and Rezaei89). Lower serum Zn2+ levels and copper toxicity [low Zn2+ to Cu2+ ratio of 0·618 (95% CI: 0·32–0·99)] were reported in ASD-C in the USA(Reference Faber, Zinn, Kern and Skip Kingston90) and in the Jilin Province, China, with a ratio of 0·64 (95% CI: 0·52–0·75) in ASD-C v. 0·74 (95% CI: 0·59–0·88) in TD children, (p = 0·000)(Reference Feng, Shan, Miao, Xue, Yue and Jia97). Those with lower [Zn2+/Cu2+] ratios fared higher on the Autism Behavior Checklist severity scale(Reference Feng, Shan, Miao, Xue, Yue and Jia97). Similarly, Zn2+ levels and the Zn2+/Cu2+ ratio were significantly lower ([1.36 × 10−5 mol/L v. 1.44 × 10−5 mol/L, p = 0·032] and [0·66 ± 0·02 v. 0·76 ± 0·02, p = 0·002], respectively) in 2–9-year-old Bangladeshi ASD-C(Reference Siddiqi, Begum, Shahjadi, Afroz, Mahruba and Parvin98); however, Cu2+ levels (p = 0·020) and ceruloplasmin levels in ASD-C were significantly higher (2.36 × 10−6 mol/L v. 2.20 × 10−6 mol/L, p = 0·045) than that of TD children. On the basis of the Chi-squared results, ASD was significantly associated with Zn2+ deficiency and Cu2+ toxicity (χ 2 = 59·239, p = 0·000 and χ 2 = 4·313, p = 0·05, respectively)(Reference Siddiqi, Begum, Shahjadi, Afroz, Mahruba and Parvin98). However, serum Zn2+ levels obtained from a large cohort of Irish children were mostly in the normal range, with no significant mean difference [0·06 μmol/L (95% CI: −0·56 to 0·67)] between participants with ASD (11·68 ± 1·7 μmol/L) and controls (11·63 ± 2·1 μmol/L)(Reference Sweetman, O’Donnell, Lalor, Grant and Greaney32). Similarly, nonsignificant differences in plasma Zn2+ levels (1.61 × 10−2 mol/L) were reported in Brazilian ASD-C(Reference Saldanha Tschinkel, Bjørklund, Conón, Chirumbolo and Nascimento99) as typical reference values are 1.10 × 10−2 – 1.76 × 10−2 mol/L(Reference Alves, de Brito, Vermeulen, Dantas Lopes, França and Bruno91). These discrepancies may be either geographically dependent, with different levels of Zn2+ in the soil(Reference Hawari, Eskandar and Alzeer100), or due to limitations in using plasma/serum concentrations as reliable indicators of Zn2+ deficiency. The sensitivity and specificity of plasma samples are limited owing to fluctuations in concentrations resulting from fasting, brief abnormal food intake, circadian variations, and inflammation(101,Reference Wieringa, Dijkhuizen, Fiorentino, Laillou and Berger102) . Moreover, most Zn2+ circulates in the blood bound to albumin. Thus, low blood levels of albumin also reduce the plasma and serum concentrations of Zn2+(Reference Wieringa, Dijkhuizen, Fiorentino, Laillou and Berger102,Reference Roohani, Hurrell, Kelishadi and Schulin103) . Alternatively, the assessment of Zn2+ levels using hair samples offers a more reliable indication of long-term Zn2+ imbalance compared with whole blood or plasma(Reference Lowe, Fekete and Decsi104,Reference DiBaise and Tarleton105) . A cross-sectional study in Italy found low levels of Zn2+ in the hair, correlating negatively with ASD severity as categorized by the behavioral scales of play and creativity (r = −0·39; p = 0·006)(Reference Fiore, Barone, Copat, Grasso, Cristaldi and Rizzo106). Similarly, another study found a significant negative correlation between Zn2+ levels and fear and nervousness (r = −0·345; p = 0·022) and between Zn2+ levels and verbal communication (r = −0·359; p = 0·017)(Reference Blaurock-Busch, Amin, Dessoki and Rabah107). Zn2+ levels in the hair were significantly lower in Asian individuals with ASD (n = 236) than those of controls (n = 306), with a mean difference of −1·493 (95% CI: −2·43 to −0·56; p = 0·002). Zn2+ concentrations in the hair of 29·7% ASD-C in Japan were below −2 standard deviations of the control reference range (86·3–193 parts per million [ppm])(Reference Yasuda, Yoshida, Yasuda and Tsutsui108). However, among non-Asian participants, the Zn2+ levels in hair were significantly higher in individuals with ASD (n = 257) than in the controls (n = 258), with a mean difference of 10·384 (95% CI: 0·04–20·72; p = 0·049)(Reference Saghazadeh, Ahangari, Hendi, Saleh and Rezaei89). Thus, inconsistent results have been reported. A recent review of studies investigating Zn2+ level disparities between individuals with ASD and TD controls concluded that significant differences in the Zn2+ concentrations in blood samples (serum and plasma) were identified, and no such disparities were found in hair or urine samples(Reference Kaczmarek, Dobrzyńska and Drzymała-Czyż109). As mentioned above, Zn2+ status is often linked to Cu2+ status, where a deficiency in one can lead to toxicity in the other, both affecting human neurodevelopmental and physiological functioning(Reference Bjørklund110,Reference Baj, Flieger, Flieger, Forma, Sitarz and Skórzyńska-Dziduszko111) . Overall, a low Zn2+/Cu2+ ratio is prevalent in ASD(Reference Bölte, Girdler and Marschik78,Reference Faber, Zinn, Kern and Skip Kingston90,Reference Li, Wang, Bjørklund, Zhao and Yin92,Reference Crăciun, Bjørklund, Tinkov, Urbina, Skalny and Rad93,Reference Bjørklund110Reference Ramaekers, Sequeira, Thöny and Quadros112) and has been proposed as a candidate diagnostic biomarker for ASD(Reference Faber, Zinn, Kern and Skip Kingston90,Reference Li, Wang, Bjørklund, Zhao and Yin92) . Collectively, a number of studies suggested that Zn2+ levels along with Zn2+/Cu2+ ratios are altered in ASD, although some inconsistencies have also been reported. Therefore, a comprehensive understanding of the role of different factors causing Zn2+ deficiency could be valuable in establishing a correlation between Zn2+ dyshomeostasis and ASD.

Role of zinc in gut physiology

Approximately 1·5–3 g of Zn2+ is present in the human body, mostly in its dissociated form(Reference Gropper, Smith and Carr113). Zn2+ is mainly stored in the skeletal muscles, bone, liver, and skin(Reference To, Do, Cho and Jung114,Reference Ross, Hernandez-Espinosa and Aizenman115) . However, it is also found in the brain, where its abundance is second only to iron in terms of essential trace minerals(Reference Ross, Hernandez-Espinosa and Aizenman115Reference DeBenedictis, Raab, Ducie, Howley, Feldmann and Grabrucker117). Therefore, it is not surprising that Zn2+ dyshomeostasis in the brain could result in neurodevelopmental and mood disorders(Reference Ross, Hernandez-Espinosa and Aizenman115,Reference Gower-Winter and Levenson118) . Zn2+ cannot be stored in the human body beyond the body’s needs. Therefore, daily dietary Zn2+ intake is essential to maintain appropriate levels in the body. Rich dietary sources of Zn2+ include oysters, red meat, and poultry from animal sources, and whole grains, beans, and nuts from plant sources(119). However, the presence of dietary phytate hinders Zn2+ absorption because the phosphate group forms strongly insoluble complexes with Zn2+, rendering it biologically unavailable(Reference Lönnerdal120). Indeed, the average Zn2+ absorption was reduced from 50% in highly refined diets to approximately 15% in unrefined diets(Reference Roohani, Hurrell, Kelishadi and Schulin103). On average, Zn2+ absorption is approximately 33%(Reference Roohani, Hurrell, Kelishadi and Schulin103); however, it can increase to 90% in cases of limited Zn2+ availability(Reference Hara, Takeda, Takagishi, Fukue, Kambe and Fukada15). Fig. 2 illustrates Zn2+ processing in the human gastrointestinal (GI) system and highlights the factors that influence Zn2+ absorption(Reference Maret and Sandstead14,Reference Lönnerdal120) .

Figure 2. Zn2+ metabolism in the human body. (a) Release of Zn2+ from nucleic and amino acid-bound complexes from food sources. (b) Zn2+ absorption through an enterocyte from the apical side through (1) ZIP4, (2) other routes, or (3) paracellular absorption. Zn2+ release from the enterocyte to the circulation occurs through ZnT1 transporter. In the blood, Zn2+ is carried by albumin (∼70%), macroglobulin (∼30%), or transferrin (∼10%). In the intracellular compartment, Zn2+ may be stored as part of metallothionein, which increases in Zn2+ supplementation and decreases with deficiency. (c) Zn2+ excretion through (1) feces, or (2) other routes.

Zn2+ absorption and secretion are controlled by two different transporter families: the zinc importer family (ZIP) and the zinc transporter family (ZnT)(Reference Marger, Schubert and Bertrand116). The mammalian genome encodes fourteen ZIP and nine ZnT transporters(Reference Hara, Takeda, Takagishi, Fukue, Kambe and Fukada15). The ZIPs are influx transporters that transport Zn2+ from extracellular fluids or intracellular compartments into the cytosol, whereas the ZnTs mostly act as efflux transporters and reduce cytosolic Zn2+ levels(Reference Hara, Takeda, Takagishi, Fukue, Kambe and Fukada15). Zn2+ is absorbed throughout the small intestine, primarily in the duodenum and jejunum(Reference Maares and Haase121). ZIP4, which is located in the apical membrane of the enterocytes, is the main transporter involved in the uptake of Zn2+ from the lumen of the gut(Reference Fukada, Yamasaki, Nishida, Murakami and Hirano17). The primary mode of intestinal Zn2+ absorption is carrier-mediated transport mechanisms(Reference Roohani, Hurrell, Kelishadi and Schulin103), which are mediated by the ZRT and IRT-like protein 4 (ZIP4), encoded by the SLC39A4 gene(Reference Jeong and Eide122). ZIP4 is regulated by dietary Zn2+ intake, where it undergoes rapid degradation with high Zn2+ intake to downregulate its absorption(Reference Hashimoto and Kambe123). ZIP4-mediated regulation is essential for maintaining Zn2+ homeostasis with minimal contributions from other carrier proteins, including divalent metal transporter-1, amino acid transporters, and ZIP11, particularly under conditions of Zn2+ restriction and paracellular diffusion(Reference Hashimoto and Kambe123,Reference Martin, Aydemir, Guthrie, Samuelson, Chang and Cousins124) . In particular, ZIP11 expression is significantly increased in the colonic tissues to enhance absorption efficiency(Reference Martin, Aydemir, Guthrie, Samuelson, Chang and Cousins124). In enterocytes, metallothionein acts as both a transient Zn2+ storage protein and a carrier to Zn2+-requiring enzymes(Reference Meguid, Bjørklund, Gebril, Doşa, Anwar and Elsaeid125). Both metallothionein and Zn2+ transporter 1 (ZnT1) control the release of Zn2+ in the portal circulation from duodenal and jejunal cells(Reference Cousins126). ZnT1 is located in the basolateral membrane of enterocytes and acinar cells, and represents the major transporter controlling the efflux of ions toward the portal circulation(Reference Cousins126). Urinary and surface loss of Zn2+ via sweat represents other routes of Zn2+ excretion(Reference Roohani, Hurrell, Kelishadi and Schulin103), both of which are adjusted during periods of Zn2+ depletion or abundance(Reference Milne, Canfield, Mahalko and Sandstead127,Reference Solomons and Caballero128) . The expression of Zn2+ transporters is influenced by various factors, and their malfunction causes Zn2+ dyshomeostasis and progression of related conditions(Reference Kambe, Tsuji, Hashimoto and Itsumura16). For example, chronic ethanol exposure alters hepatic Zn2+ transporters via oxidative stress, ultimately leading to decreased Zn2+ levels in the liver(Reference Sun, Li, Zhong, Zhang, Sun and Tan129). In mouse and human pancreatic islet cells, hypoxia decreases the expression of the Zn2+ transporter 8 and cytosolic Zn2+ levels(Reference Gerber, Bellomo, Hodson, Meur, Solomou and Mitchell130). Several hormones and cytokines play roles in maintaining systemic and cellular Zn2+ homeostasis. Melatonin regulates the levels of the Zn2+ transporters in the duodenal, jejunal, and ileal tissues (ZnT2, ZIP2, and ZIP4, respectively) to enhance Zn2+ absorption(Reference Baltaci and Mogulkoc131,Reference Unal, Baltaci, Mogulkoc and Avunduk132) . Hepcidin is a regulator of iron metabolism; however, it significantly reduces the availability of cellular ZnT1, thereby inhibiting Zn2+ export(Reference Hennigar and McClung133). Estrogen modulates the expression of ZnT3 and Zn2+ in hippocampal synapses(Reference Lee, Kim, Hong, Lee, Cherny and Bush134). Exposure to pro-inflammatory cytokines (particularly interleukin (IL)-1β) down-regulates Zn2+ transporters in pancreatic islet cells(Reference Egefjord, Jensen, Bang-Berthelsen, Petersen, Smidt and Schmitz135,Reference Muayed, Billings, Raja, Zhang, Park and Newman136) . Other conditions, such as infection and trauma, may disrupt the Zn2+ equilibrium, whereby Zn2+ is shifted into cellular compartments due to increased demand for protein synthesis and neutralize the free radicals(Reference Gammoh and Rink137). Zn2+ levels are also affected by conditions that alter serum albumin levels, including inflammation-related albumin decline(Reference Roohani, Hurrell, Kelishadi and Schulin103,Reference King138) .

Zn2+ deficiency enhances ZIP4 expression in the intestine. Following a 1 day Zn2+-deficient diet, ZIP4 showed a more rapid accumulation rate in the jejunum than in the duodenum(Reference Hashimoto, Nakagawa, Tsujimura, Miyazaki, Kizu and Goto139). However, this adaptive response also increases the uptake of toxic metals, such as cadmium and lead, which may contribute to the severity of ASD(Reference Yasuda, Yoshida, Yasuda and Tsutsui108). Low levels of Zn2+ have been associated with elevated lead levels(Reference Wani, Ansari, Ahmad, Parveen, Siddique and Shadab140), and lead retention in the brain affects the behavioral performance of animals(Reference Bushnell and Levin141). Similarly, Zn2+ showed a significant inverse association with serum Cadmium concentration. Specifically, a 10% increase in serum Zn2+ levels was associated with an approximately 2% decrease in blood cadmium (95% CI: −3·17 to −0·81) and an approximately 4% increase in urinary cadmium (95% CI: 2·14–6·04)(Reference Vance and Chun142). Thus, disturbances in Zn2+ levels are associated with imbalances in other minerals, indicating the significance of Zn2+ homeostasis in gut and brain physiology.

Role of zinc in microbial physiology

Metal ions, including Zn2+, are essential for the survival of all microbes, since they are required for several biological processes and act as cofactors for many enzymes. It is critical that the uptake of metal ions is in accordance with their metabolic needs because an imbalance in metal homeostasis has deleterious effects on bacteria(Reference Porcheron, Garenaux, Proulx, Sabri and Dozois143). Bacterial cells need to achieve a fine-tuned balance between ensuring sufficient concentrations of Zn2+ to fulfill essential functions and limiting their concentration to prevent toxic effects(Reference Porcheron, Garenaux, Proulx, Sabri and Dozois143).

Zn2+ transport across the outer bacterial membrane is not defined in Enterobacteria, which represents an important component of the gut microbiota; however, Zn2+ uptake across the cytoplasmic membrane is mediated by two major types of transporters, a high affinity transporter “ZnuACB” and a low-affinity uptake system “ZupT,” which are expressed under conditions of low and moderate Zn2+ availability, respectively(Reference Porcheron, Garenaux, Proulx, Sabri and Dozois143). Studies have demonstrated that mutations in the ZnuACB genes can decrease Zn2+ uptake in Enterobacteria(Reference Gabbianelli, Scotti, Ammendola, Petrarca, Nicolini and Battistoni144,Reference Nielubowicz, Smith and Mobley145) . In some bacterial species, such as Escherichia coli (O157:H7) and Salmonella Typhimurium (S. Typhimurium), Zn2+ uptake involves another protein, ZinT (formerly known as YodA), which mediates periplasmic Zn2+ binding under Zn2+-limiting conditions(Reference Gabbianelli, Scotti, Ammendola, Petrarca, Nicolini and Battistoni144,Reference Bersch, Bougault, Roux, Favier, Vernet and Durmort146) .

ZnuACB contributes to resistance against host calprotectin-mediated Zn2+ chelation(Reference Gabbianelli, Scotti, Ammendola, Petrarca, Nicolini and Battistoni144,Reference Liu, Jellbauer, Poe, Ton, Pesciaroli and Kehl-Fie147) , which accumulates in the host intestine after infection(Reference Liu, Jellbauer, Poe, Ton, Pesciaroli and Kehl-Fie147). Moreover, pathogenic bacteria, such as S. Typhimurium, exploit calprotectin-mediated Zn2+ chelation to outcompete the host microbiota, which is less well adapted to the Zn2+-limited environment in the infected intestine(Reference Liu, Jellbauer, Poe, Ton, Pesciaroli and Kehl-Fie147). This study showed that Zn2+ availability in the inflamed gut is an important means of growth and competition between microbes. It is evident that pathogens compete with the resident microbiota for nutrients by acquiring specialized uptake systems. Some species of commensal microbes in the gut utilize metal transporters to grow under metal-limiting conditions(Reference Liu, Jellbauer, Poe, Ton, Pesciaroli and Kehl-Fie147).

Effect of zinc on gut microbiota

Zn2+ is an essential trace element for gut commensal microbiota and plays a significant role in the normal functioning of multiple organs and systems in the human body, particularly the immune and endocrine systems. Furthermore, the role of the gut microbiota in regulating the gut–brain axis linked to several neuropsychiatric and neurodegenerative disorders, including ASD, has been proven to be important(Reference Shoubridge, Choo, Martin, Keating, Wong and Licinio148). However, data on the relationship between GI tract diseases, the gut microbiota, and Zn2+ transporters in individuals with ASD are limited.

The gut microbiota can bind mineral particles and pathogens and compete with commensals and enterocytes for Zn2+ absorption. Zn2+ can affect the balance between pathogenic and commensal strains(Reference Scarpellini, Balsiger, Maurizi, Rinninella, Gasbarrini and Giostra22). Although Zn2+ is an essential micronutrient for bacteria, its excess results in significant toxicity to bacteria(Reference McDevitt, Ogunniyi, Valkov, Lawrence, Kobe and McEwan149). Zn2+ overexposure has been shown to significantly alter gut microbiota, promoting a shift towards harmful bacteria, including pathogenic Escherichia coli varieties. This is likely due to its high bioavailability, which supports the growth of pathogenic bacteria(Reference Skalny, Aschner, Lei, Gritsenko, Santamaria and Alekseenko150). Animal studies have shown that long- and short-term feeding of mice diets supplemented with Zn2+ is associated with alterations in gut bacterial composition at the phylum, genus, and species levels(Reference Chen, Wang, Wang, Yu, Ding and Wang151). Low, normal, high, and excessive amounts of Zn2+ (0, 30, 150, and 600 mg/kg Zn2+) were fed to 3-week-old C57BL/6 mice and maintained for either four weeks (short-term Zn2+ intervention) or eight weeks (long-term Zn2+ intervention) to demonstrate its effects on various developmental stages from birth to puberty(Reference Chen, Wang, Wang, Yu, Ding and Wang151). Analysis of the relative abundances of the dominant genera revealed distinct clustering of cecal microbiota in mice fed diets containing various levels of Zn2+. During short-term Zn2+ intervention, the microbiota of mice fed with a low-Zn2+ diet was characterized by enrichment of Desulfovibrio, Enterorhabdus, Flavonifractor, Parasutterella, Mucispirillum, Candidatus Saccharimonas, Alistipes, Odoribacter, and Anaerotruncus, and a decrease in the relative abundance of Sphingomonas (Reference Chen, Wang, Wang, Yu, Ding and Wang151). A similar pattern has been reported in individuals with ASD, where Desulfovibrio, Flavonifractor, and Alistipes were enriched(Reference Kittana, Ahmadani, Al Marzooq and Attlee152). The relative abundances of unidentified Lachnospiraceae, Lachnoclostridium, and Bifidobacterium in the control-Zn2+ group were higher than those in the high- and excess-Zn2+ groups(Reference Chen, Wang, Wang, Yu, Ding and Wang151). All these bacteria have a protective role in the gut but have been found to be depleted in the ASD-C group(Reference Ma, Liang, Dai, Wang, Luo and Zhang153). The high- Zn2+ diet was associated with the enrichment of Akkermansia, Faecalibaculum, Helicobacter, and Ileibacterium compared with other diets(Reference Chen, Wang, Wang, Yu, Ding and Wang151). Mice fed with a 600 mg/kg Zn2+ diet showed a higher abundance of Dubosiella, Caulobacter, and Bradyrhizobium, and a lower proportion of Romboutsia, Bacteroides, Lactobacillus, and Bifidobacterium (Reference Chen, Wang, Wang, Yu, Ding and Wang151). These findings contrasted with typical ASD microbiota profiles, where Akkermansia is usually depleted and Lactobacillus is enriched, suggesting the protective role of a high Zn2+ diet(Reference Xu, Xu, Li and Li154). In addition, the observed high levels of Helicobacter (Reference Bougafa155) and low levels of beneficial Bacteroides and Bifidobacterium were consistent with known ASD gut microbiota imbalances(Reference Xu, Xu, Li and Li154). Notably, during short-term intervention, the ratio of Bacteroidetes to Firmicutes was negatively correlated with the Zn2+ dosage(Reference Chen, Wang, Wang, Yu, Ding and Wang151). A low Bacteroidetes:Firmicutes ratio is a typical feature of ASD(Reference Kittana, Ahmadani, Al Marzooq and Attlee152).

Long-term Zn2+ intervention, compared with short-term Zn2+ intervention, reversed the distribution of some bacterial genera and led to an increase in the relative abundance of Akkermansia, Blautia, Alloprevotella, and Ruminiclostridium, and a decrease in Thermovirga (Reference Chen, Wang, Wang, Yu, Ding and Wang151). The relative abundances of Parasutterella, Helicobacter, Odoribacter, and Ileibacterium in the control-Zn2+ group were higher, and the proportions of Dubosiella, Faecalibaculum, and Bifidobacterium were lower than those in the other groups(Reference Chen, Wang, Wang, Yu, Ding and Wang151). In contrast, long-term high- Zn2+ intervention led to a decrease in most genera compared with the other dietary treatments, except for increased levels of Bifidobacterium and Anaeroplasma. Excess Zn2+ diet-fed mice showed increased levels of Bacteroides and Intestinimonas and decreased levels of Lactobacillus (Reference Chen, Wang, Wang, Yu, Ding and Wang151). Moreover, pathways, including carbohydrate, glycan, and nucleotide metabolism, were decreased by a short-term low-Zn2+ diet. Long-term Zn2+ use, especially at high-Zn2+ doses, suppresses the abundance of short-chain fatty acid (SCFA)-acid-producing genera and their metabolites(Reference Chen, Wang, Wang, Yu, Ding and Wang151).

Recently, Zn2+ exposure was found to alter gut microbiota composition in an in vitro simulation of the human colon, whereby exposure to nanoparticles of zinc oxide (ZnO) led to dose-dependent changes in the composition and key functional pathways controlled by the gut microbiome(Reference Zhang, Zhu, Guo, Gu, Li and Chen156). The relative abundance of Bacteroidetes was high, whereas that of Firmicutes was low. Bacterial biodiversity, SCFA production, and antibiotic resistance genes were also reduced(Reference Zhang, Zhu, Guo, Gu, Li and Chen156). However, the diversity of the gut microbiota recovered after ZnO nanoparticle exposure was discontinued(Reference Zhang, Zhu, Guo, Gu, Li and Chen156).

Role of zinc in maintaining a healthy gut

Zn2+ contributes to GI development, and its deficiency is involved in the etiology of a wide range of GI diseases(Reference Skrovanek, DiGuilio, Bailey, Huntington, Urbas and Mayilvaganan157). Overall, the evidence indicates that Zn2+ is essential for maintaining proper intestinal function and is a significant factor in gut microbiota biodiversity(Reference Skalny, Aschner, Lei, Gritsenko, Santamaria and Alekseenko150). In addition, Zn2+ can modulate gut microbiota to promote the integrity of the intestinal barrier, reduce inflammation, aid the regeneration of the epithelium, and control the permeation of pathogens(Reference Xia, Lian, Wu, Yan, Quan and Zhu158). Fig. 3 shows the interplay between gut microbiota and Zn2+ in maintaining a healthy gut. Inhibition of bacterial translocation into systemic circulation is associated with maintenance of adequate expression of intestinal tight junction (TJ) proteins(Reference Ulluwishewa, Anderson, McNabb, Moughan, Wells and Roy159).

Figure 3. Interplay between gut microbiota and Zn2+ to maintain a healthy gut. (a) In a Zn2+-abundant state, bacteria of the following phyla seem to thrive: Firmicutes, Actinobacteria, Verrucumicrobia, Proteobacteria, and Bacteroidetes. (b) In addition, the mucous membranes are intact with healthy enterocytes, and TJ. No inflammation or bacterial penetration is found. (c) In a Zn2+-deficient state, a decrease in Thermovirga and increase in Akkermansia, Blautia, Alloprevotella, and Ruminiclostridium were found in the cecum of mice, with an enrichment of Helicobacter hepaticus. (d) Enterocyte inflammation, impaired TJ, and proliferation of neutrophils along with pathogenic bacteria are found in a Zn2+-deficient state.

Zn2+ critically regulates TJ complexes in the gut epithelium(Reference Ulluwishewa, Anderson, McNabb, Moughan, Wells and Roy159). The epithelial and endothelial TJ selectively seal the space between the adjacent cells, preventing unregulated paracellular exchange across the epithelial and endothelial barriers(Reference Ulluwishewa, Anderson, McNabb, Moughan, Wells and Roy159). Exposure to pathogenic bacteria, certain foods, or micronutrients can drastically affect the ability of the TJ to regulate the permeation of nutrients, water, and electrolytes(Reference Ulluwishewa, Anderson, McNabb, Moughan, Wells and Roy159). Several studies have shown that Zn2+ supplementation or deprivation affects the epithelial barrier function, which is attributed to Zn2+-mediated TJ modifications(Reference Zhong, McClain, Cave, Kang and Zhou160).

Zn2+ deficiency can cause intestinal hyperpermeability, commonly known as leaky gut, allowing toxins and antigens produced by harmful bacteria to cross the intestinal lumen and enter the bloodstream(Reference Al-Ayadhi, Zayed, Bhat, Moubayed, Al-Muammar and El-Ansary161). Recent studies have reported that probiotic bacteria, such as Bifidobacterium and Lactobacillus, can enhance the production of TJ proteins, thus reversing leaky gut disorders(Reference Al-Ayadhi, Zayed, Bhat, Moubayed, Al-Muammar and El-Ansary161). Therefore, maintaining a normal gut microbiota is critical for the development of an effective intestinal barrier(Reference Al-Ayadhi, Zayed, Bhat, Moubayed, Al-Muammar and El-Ansary161).

Zinc status, altered gut microbiota, and ASD

Alterations in GI microbiota have frequently been reported in ASD. Several studies have indicated that GI pro-inflammatory factors, including bacterial lipopolysaccharides (LPS), can induce an inflammatory response that eventually affects brain development(Reference Kirsten, Queiroz-Hazarbassanov, Bernardi and Felicio162,Reference Kalyan, Tousif, Sonali, Vichitra, Sunanda and Praveenraj163) . Leaky guts have often been reported in ASD(Reference Al-Ayadhi, Zayed, Bhat, Moubayed, Al-Muammar and El-Ansary161,Reference Navarro, Pearson, Fatheree, Mansour, Hashmi and Rhoads164Reference Fiorentino, Sapone, Senger, Camhi, Kadzielski and Buie167) . It is associated with a reduced abundance of probiotic bacteria, overgrowth of pathogenic bacteria, accumulation of toxic metabolites, and release of pro-inflammatory cytokines, leading to the development of autistic features through alteration of the gut–brain axis(Reference Al-Ayadhi, Zayed, Bhat, Moubayed, Al-Muammar and El-Ansary161), as postulated in Fig. 4.

Figure 4. Crosstalk between microbiota-derived metabolites, their role in altering brain activity, and the potential impact of Zn2+ on the entire process (a, b). SCFAs may reach the CNS through the vagus nerve and play a neuroactive role (b). (c) Accumulation of toxic metabolites (lipopolysaccharides) produced by distinct gut bacteria that promote immune cell recruitment (d) and trigger the release of pro-inflammatory cytokines (e), could be key factors in the development of autistic phenotypes by affecting the synapses in the brain (f) via the gut–brain axis. (g) Zn2+ promotes neural communication and reduces inflammation by suppressing immune cell recruitment and reducing the production of pro-inflammatory cytokines. By contrast, low Zn2+ status can compromise intestinal barrier integrity and activate pro-inflammatory signaling, resulting in changes in microbiota composition that may aggravate inflammation.

In general, there are few studies on the potential association between Zn2+ status and gut microbiota in humans. Previous studies have demonstrated that Zn2+ deficiency is associated with reduced gut microbiota biodiversity(Reference Sauer and Grabrucker168Reference Chen, Jiang, Wang, Chen, Tang and Wang170). However, unique taxa, associated with Zn2+ deficiency, could not be identified because of limited data available from human studies(Reference Skalny, Aschner, Lei, Gritsenko, Santamaria and Alekseenko150). Restricted access to Zn2+ may influence the maternal microbiota composition and, subsequently, its composition in human offspring(Reference Sauer and Grabrucker168). Furthermore, the offspring of mice with Zn2+ deficiency during pregnancy exhibited ASD-like behavior(Reference Vela, Stark, Socha, Sauer, Hagmeyer and Grabrucker36,Reference Sauer, Hagmeyer and Grabrucker171Reference Schoen, Asoglu, Bauer, Müller, Abaei and Sauer174) . Interestingly, low dietary levels or bioavailability of Zn2+ are associated with altered microbiota composition and inflammation in pregnant mice, establishing a link between maternal Zn2+ deficiency, altered microbiota composition, and inflammation(Reference Sauer and Grabrucker168). Consequently, inflammatory processes, such as neuroinflammation, which can arise under altered microbiota composition, may contribute to altered brain development in ASD(Reference Sauer and Grabrucker168). A recent study showed that Zn2+ is a key regulator of GI development, microbiota composition, and inflammation, with relevance in ASD, and indicated that maternal Zn2+ deficiency in mice affects the neonatal Zn2+ status of the offspring(Reference Sauer, Malijauskaite, Meleady, Boeckers, McGourty and Grabrucker175). In a recent meta-analysis comparing biomedical factors, trace elements, and microbiota biomarkers between children and adolescents with ASD and TD controls, significant differences were observed. Specifically, ASD-C displayed significantly lower Zn2+ levels (mean difference = −6·707, 95% CI: −12·691 to −0·722) and altered microbiota composition, including decreased relative abundance of Bifidobacterium (MD = −1·321, 95% CI: −2·403 to −0·238) and Parabacteroides (MD = −0·081, 95% CI: −0·148 to −0·013), alongside elevated levels of Bacteroides (MD = 1·386, 95% CI: 0·717–2·055) and Clostridium (MD = 0·281, 95% CI: 0·035–0·526)(Reference Lin, Zhang, Sun, Li, Li and Zhu176). Proteomic approaches have revealed that Zn2+ deficiency affects several biological processes, including altered GI physiology and pro-inflammatory signaling, resulting in chronic systemic inflammation, neuroinflammation, and abnormal gut microbiota composition similar to that reported in ASD cases in human subjects(Reference Sauer, Malijauskaite, Meleady, Boeckers, McGourty and Grabrucker175). The microbiota composition of mice with prenatal Zn2+ deficiency was significantly altered, with an increase in the phyla Actinobacteria, Proteobacteria, and Tenericutes, and a decrease in Firmicutes(Reference Sauer and Grabrucker168). Overall, it appears that a low maternal Zn2+ status during fetal development is sufficient to compromise intestinal barrier integrity and activate pro-inflammatory signaling, resulting in changes in microbiota composition that may aggravate inflammation, mimicking the comorbidities frequently observed in ASD(Reference Sauer and Grabrucker168,Reference Sauer, Malijauskaite, Meleady, Boeckers, McGourty and Grabrucker175) . Thus, alterations in microbiota composition combined with abnormal GI physiology and/or morphology may trigger inflammatory reactions. However, the mechanistic links among inflammation, GI and brain abnormalities, and maternal Zn2+ status in individuals with ASD remain unclear.

Role of zinc in brain neurophysiology

Zn2+ is a potent neuromodulating agent that functions as a signaling ion, promotes neuronal and glial cell development, and regulates the levels of the neurotrophin, brain-derived neurotrophic factor (BDNF)(Reference Kirsten, Queiroz-Hazarbassanov, Bernardi and Felicio162). Therefore, Zn2+ homeostasis controlled by Zn2+ transporters in this complex organ is crucial(Reference Benedictis, Haffke, Hagmeyer, Sauer and Grabrucker177). Dysfunction of these transporters appears to play a role in the etiology of ASD(Reference Benedictis, Haffke, Hagmeyer, Sauer and Grabrucker177,Reference Levaot and Hershfinkel178) . For example, male ZnT3 null mice, as opposed to females, exhibited ASD-like behavior, and ablation of the ZnT3 transporter altered synaptic and cytosolic Zn2+, elevating the activity of BDNF(Reference Yoo, Kim, Yoon and Koh179). ZIP4 transporters are most prevalent in neurons that control excitatory synapses, and are sensitive to short-term changes in Zn2+ levels at the synapses(Reference Benedictis, Haffke, Hagmeyer, Sauer and Grabrucker177). In the brain, Zn2+ regulates gene expression through the Zn2+ finger transcription factors, thereby influencing neurogenesis and region-specific brain development(Reference Marger, Schubert and Bertrand116). Zn2+ is released from the presynaptic terminals and binds to the receptors, ion channels, and transporters, thus modulating synaptic transmission and plasticity(Reference Kambe, Tsuji, Hashimoto and Itsumura16). Approximately 10% of the Zn2+ in the CNS is not ligand-associated and is found in the presynaptic vesicles of the glutamatergic neurons(Reference Gower-Winter and Levenson118,Reference Pochwat, Nowak and Szewczyk180,Reference Frederickson, Suh, Silva, Frederickson and Thompson181) in the cerebral cortex, amygdala, dorsal cochlear nucleus, and hippocampus(Reference Krall, Moutal, Phillips, Asraf, Johnson and Khanna182). It has been demonstrated that the ZnT3 loads the presynaptic vesicles with Zn2+, and its expression is important for maintaining Zn2+ homeostasis and hippocampus-dependent learning, which are disrupted in ZnT3 knockout mice(Reference Sindreu, Palmiter and Storm183). The mossy fibers from the hippocampal cornu ammonis (CA3) area contain one of the highest concentrations of Zn2+ in the brain, which are assumed to be co-released with glutamate via exocytosis(Reference Li, Hough, Frederickson and Sarvey184Reference Quinta-Ferreira, Sampaio dos Aidos, Matias, Mendes, Dionísio and Santos186). After its release into the synaptic cleft and diffusion, the hippocampal mossy fiber Zn2+ may modulate different pre- and post-synaptic mechanisms(Reference Li, Hough, Frederickson and Sarvey184Reference Quinta-Ferreira, Sampaio dos Aidos, Matias, Mendes, Dionísio and Santos186). In particular, voltage-gated potassium (K+) channels (Kv1·1 and Kv1·4) are expressed at the mossy fiber terminals and control crucial neuronal functions, such as excitability, action potential shape, and neurotransmitter release(Reference Assaf and Chung187Reference Wang, Kunkel, Schwartzkroin and Tempel189). It has been shown that these channel types are inhibited by Zn2+ and that Zn2+ facilitates synaptic transmission in the hippocampus by depressing the activity of the pre-synaptic delayed-rectifier K+ channels composed of both the Kv1·1 and Kv1·4 subunits(Reference Cusimano, D’Adamo and Pessia190,Reference Imbrici, D’Adamo, Cusimano and Pessia191) . The N-methyl-d-aspartate receptor (NMDAR) is a postsynaptic receptor with high affinity for Zn2+(Reference Krall, Moutal, Phillips, Asraf, Johnson and Khanna182). The NMDAR modulates excitatory transmission stimulated by glutamate and plays an essential role in synaptic transmission and plasticity(Reference Petrilli, Kranz, Kleinhaus, Joe, Getz and Johnson192,Reference Amico-Ruvio, Murthy, Smith and Popescu193) . The NMDAR are allosterically inhibited by Zn2+, which reduces NDMAR activity by stimulating the release of the neurotransmitter GABA, which exerts an inhibitory effect on glutamate release at the presynaptic level(Reference Petrilli, Kranz, Kleinhaus, Joe, Getz and Johnson192). Events leading to increased postsynaptic Zn2+ levels activate several signaling pathways that exert neurotoxic effects and cause cell death(Reference Pochwat, Nowak and Szewczyk180). Zn2+ plays a role in neurotoxicity in models of brain-related pathologies, such as seizures, which are a comorbidity often observed in ASD(Reference Pochwat, Nowak and Szewczyk180). Overall, this evidence highlighted the importance of Zn2+ in maintaining proper neuronal and synaptic functions and supported the notion that Zn2+ dyshomeostasis could be a critical risk factor for ASD.

Zinc transporters—SHANK3 crosstalk in ASD

There exists a crosstalk between ZIP4 and SHANK. The SHANK proteins are encoded by SHANK1, SHANK2, and SHANK3 and comprise the multidomain scaffold proteins at the postsynaptic density of glutamatergic synapses. These proteins connect the neurotransmitter receptors, ion channels, and other membrane proteins to the actin cytoskeleton and G protein-coupled signaling pathways. SHANK also plays a role in synaptic formation and dendritic spine maturation in the brain(Reference Shi, Redman, Ghose, Hwang, Liu and Ren194). Mutations in SHANK3 on chromosome 22 are associated with ASD(Reference Uchino and Waga195). A study involving pluripotent cells derived from individuals with Phelan–McDermid syndrome (PMDS) and ASD features showed reduced SHANK3 expression(Reference Pfaender, Sauer, Hagmeyer, Mangus, Linta and Liebau196). Interestingly, decreased expression of the ZIP2 and ZIP4 transporters has also been observed, especially at SHANK3 sites, suggesting abnormal control of Zn2+ homeostasis in these individuals(Reference Pfaender, Sauer, Hagmeyer, Mangus, Linta and Liebau196). It is possible that the Zn2+ deficiency observed in the individuals with ASD decreases the expression of the SHANK3, ZIP2, and ZIP4 proteins(Reference Levaot and Hershfinkel178). Restoration of the SHANK3 levels or downstream mediators in adults may be a useful therapeutic approach for alleviating synaptic and behavioral impairments associated with SHANK3 mutations(Reference Monteiro and Feng197). As Zn2+ deficiency exhausts both Zn2+ pools and SHANK3 availability, lifelong co-therapy with Zn2+ supplementation may be required to promote scaffold formation and improve synaptic plasticity(Reference Hagmeyer, Sauer and Grabrucker65). Therefore, it could be hypothesized that individuals with PMDS and ASD caused by Shankopathies, with one intact copy of SHANK3, may benefit from Zn2+ supplementation, as elevated Zn2+ may drive the remaining SHANK3 to the postsynaptic density and additionally recruit SHANK2, a second Zn2+-dependent member of the SHANK gene family(Reference Hagmeyer, Sauer and Grabrucker65). Zn2+ may also exert its effects through direct modulation of SHANK proteins (Reference Hagmeyer, Sauer and Grabrucker65,Reference Arons, Lee, Thynne, Kim, Schob and Kindler198,Reference Grabrucker, Jannetti, Eckert, Gaub, Chhabra and Pfaender199) , as SHANK3 has a domain called the sterile alpha motif (SAM) to which Zn2+ binds. Altered Zn2+ levels in pools stored in glutamatergic synaptic vesicles may affect neurotransmitter release, postsynaptic responses, and trans-synaptic coupling(Reference Arons, Lee, Thynne, Kim, Schob and Kindler198).

The relationship between SHANK, gut microbiota, and ASD

In addition to the brain, SHANK3 is expressed in the gut epithelium, liver, heart, kidneys, and skeletal muscles(Reference Sauer, Bockmann, Steinestel, Boeckers and Grabrucker200,Reference Kim, Ko, Jin, Zhang, Kang and Ma201) . The use of ASD mouse models is crucial for understanding how microbes affect neurodevelopmental conditions and for discovering targets for the treatment of ASD(Reference Schoen, Asoglu, Bauer, Müller, Abaei and Sauer174,Reference Sauer, Bockmann, Steinestel, Boeckers and Grabrucker200,Reference Lee, Jung, Vyas, Skelton, Abraham and Hsueh202Reference Kazdoba, Leach and Crawley204) . In particular, SHANK3 knockout mice exhibit GI dysfunction, including dysbiotic gut microbiota composition(Reference Tabouy, Getselter, Ziv, Karpuj, Tabouy and Lukic205) and abnormal absorption of trace minerals, such as Zn2+(Reference Pfaender, Sauer, Hagmeyer, Mangus, Linta and Liebau196). Low enterocyte levels of SHANK3 also reduced ZIP4 gene expression, and consequently, protein expression(Reference Pfaender, Sauer, Hagmeyer, Mangus, Linta and Liebau196). SHANK3 plays an important role in gut permeability by affecting intestinal barrier function(Reference Wei, Yang-Yen, Tsao, Weng, Tung and Yu206). The SAM in the C-terminal domain of the SHANK3 proteins contains a Zn2+-binding site; thus, SHANK3 requires Zn2+ to function properly(Reference Wong, Montgomery, Taylor and Grabrucker207). The main SHANK3 rodent model that has been widely utilized in ASD research is the Shank3B−/− ex13–16 mouse, in which exons 13–16 of Shank3B−/− are deleted(Reference Wei, Yang-Yen, Tsao, Weng, Tung and Yu206,Reference Wong, Montgomery, Taylor and Grabrucker207) . This effect is similar to knocking out the gene, which causes a loss-of-function of the SHANK3 protein, as observed in human subjects with ASD. Shank3B−/− mice display the loss of several proteins in the postsynaptic structure, similar to those seen in individuals with ASD, and autistic-like-traits, including repetitive behaviors and deficits in social interaction(Reference Wong, Montgomery, Taylor and Grabrucker207).

Notably, differences in the gut microbiota of Shank3B−/− mice compared with wild-type (WT) littermates have been observed; thus, this mouse strain has been used to explore the interplay between the gut microbiota and ASD(Reference Sgritta, Dooling, Buffington, Momin, Francis and Britton208,Reference Wong, Jung, Lee, Fourie, Handley and Montgomery209) . In the same model, Zn2+ supplementation (150 ppm) was shown to reverse alterations in bacterial diversity, expression of TJ genes, and genes involved in immune regulation and energy metabolism(Reference Wong, Jung, Lee, Fourie, Handley and Montgomery209).

Probiotic treatment with Lactobacillus reuteri ameliorated ASD-like behaviors caused by dysbiosis in the gut microbiota of Shank3B−/− mice(Reference Tabouy, Getselter, Ziv, Karpuj, Tabouy and Lukic205). This was positively correlated with the expression of the GABA receptor subunits in the brains of these mice(Reference Tabouy, Getselter, Ziv, Karpuj, Tabouy and Lukic205). Studies have shown that Zn2+ supplementation was beneficial in the Shank3B−/− mouse model of ASD, wherein 6 weeks of postnatal supplementation of Zn2+ in the diet resulted in the reversal of some of the ASD-related behaviors(Reference Fourie, Vyas, Lee, Jung, Garner and Montgomery210). Moreover, Zn2+ supplementation in pregnant Shank3B−/− mice inhibited the development of ASD-related behaviors in their offspring(Reference Fourie, Vyas, Lee, Jung, Garner and Montgomery210). This clear link between dietary Zn2+ and ASD behavior suggests the possible involvement of Zn2+-dependent signaling in the gut–microbiota–brain axis in ASD(Reference Wong, Jung, Lee, Fourie, Handley and Montgomery209). A recent study explored the changes in the gut microbiota in relation to autistic-like behaviors of Shank3B−/− mice, wherein Zn2+ was supplemented in the diet (using 30 and 150 ppm Zn2+). Four types of GI samples (ileum, cecum, colon, and fecal) were collected from the WT and Shank3B−/− mice on either control or supplemented Zn2+ diets(Reference Wong, Jung, Lee, Fourie, Handley and Montgomery209). Specific microbial taxa were differentially abundant in the four experimental groups. A total of seventy-four taxa, mostly from the families Muribaculaceae (Bacteroidetes) and Lachnospiraceae (Firmicutes), were significantly and differentially represented between the WT and Shank3B−/− mice fed with Zn2+ (30 ppm)(Reference Wong, Jung, Lee, Fourie, Handley and Montgomery209). Muribaculaceae, Erysipelotrichaceae, and Blautia were more abundant in the WT mice than in the Shank3B−/− mice fed with Zn2+. Blautia is particularly important because it produces butyrate, a SCFA that is beneficial for host health owing to its anti-inflammatory effects on the mucosa. It also exerts positive neuromodulatory effects on the gut–microbiota–brain axis, owing to its ability to cross the blood–brain barrier(Reference Wong, Jung, Lee, Fourie, Handley and Montgomery209,Reference Mirzaei, Bouzari, Hosseini-Fard, Mazaheri, Ahmadyousefi and Abdi211) . SCFA may also contribute to lowering the intestinal pH, increasing Zn2+ solubility, and thus, increasing the absorption of dietary Zn2+(Reference Wong, Montgomery, Taylor and Grabrucker207,Reference Wong, Jung, Lee, Fourie, Handley and Montgomery209) . Among the Shank3B−/− mice, the bacterial Enterobacteriaceae and Coriobacteriaceae families were more abundant in those supplemented with Zn2+ diet (150 ppm) than in the control Zn2+ diet (30 ppm) mice, which had an increased abundance of Muribaculaceae and Ileibacterium (Reference Wong, Jung, Lee, Fourie, Handley and Montgomery209). Diets can alter the host production of bile acids, which can affect the gut microbiome, favoring the proliferation of the bacterial species that metabolize bile acids, such as members of the Coriobacteriaceae(Reference Wong, Jung, Lee, Fourie, Handley and Montgomery209). This, in turn, can affect gut barrier permeability through the interaction of bile acids with the epithelial cells. Similarly, members of Muribaculaceae, Blautia, and Faecalibaculum were more abundant in WT mice fed a 30 ppm Zn2+ diet compared with the Shank3B−/− mice fed with a 150 ppm Zn2+ diet, which had a higher abundance of Coriobacteriaceae and Escherichia/Shigella (Reference Wong, Jung, Lee, Fourie, Handley and Montgomery209). From the above studies, it is evident that altered Zn2+ status can cause dysbiosis by altering microbial composition and diversity. In the presence of dysbiotic microbiota, the overall microbial products change, affecting gut function and the ability to process nutrients(Reference Chen, Wang, Wang, Yu, Ding and Wang151). This evidence suggested a link between genes associated with ASD, dietary Zn2+, and gut microbiota in the pathophysiology of ASD.

Potential role of prenatal zinc deficiency in ASD

Zn2+, among other trace minerals, plays a critical role in early neurodevelopment(Reference Ross, Hernandez-Espinosa and Aizenman115,Reference Gower-Winter and Levenson118) . Various factors that direct stem cell proliferation during neurodevelopment require Zn2+(Reference Frederickson, Suh, Silva, Frederickson and Thompson181). Prenatal Zn2+ deficiency (PZD) is considered an environmental risk factor for ASD development in the offspring(Reference Grabrucker212) which has direct and indirect implications(Reference Vela, Stark, Socha, Sauer, Hagmeyer and Grabrucker36,Reference Sauer, Hagmeyer and Grabrucker171Reference Schoen, Asoglu, Bauer, Müller, Abaei and Sauer174) . Zn2+ is supplied to the brain via the blood–brain and blood–cerebrospinal fluid barriers to maintain homeostasis(Reference Takeda213). This ion is stored in the amygdala and Zn2+-abundant glutamatergic neurons, which act as the neuromodulators of synaptic transmission(Reference Takeda213). The synaptic vesicles store Zn2+ at high concentrations and regulate its release into the synaptic cleft after the physiological stimulation of the glutamatergic neurons(Reference Zhang, Dischler, Glover and Qin214). Therefore, dietary Zn2+ deprivation, particularly during early brain development, may influence Zn2+ homeostasis by affecting zincergic innervation(Reference Sauer, Hagmeyer and Grabrucker171,Reference Zhang, Dischler, Glover and Qin214) . Low maternal Zn2+ levels result in reduced brain Zn2+ levels in offspring(Reference Grabrucker, Boeckers and Grabrucker172,Reference Grabrucker, Jannetti, Eckert, Gaub, Chhabra and Pfaender199,Reference Grabrucker212,Reference Liu, Adamo and Oteiza215) . Signaling pathways at the glutamatergic synapses, involving SHANK2 and SHANK3 proteins, are influenced by PZD and linked to ASD(Reference Grabrucker212). PZD animal models, including mice born to mothers fed a 5-week Zn2+ deficient diet (< 5 ppm) prior to mating and during gestation(Reference Grabrucker212) also showed some ASD-like behaviors, such as increased anxiety levels, impaired nest building and vocalizations, and altered social interactions(Reference Hagmeyer, Haderspeck and Grabrucker31). Additional consequences included the loss of a family of proteins that have a major scaffolding function at the synapse, namely the ProSAP1/SHANK2 and ProSAP2/SHANK3 proteins(Reference Grabrucker, Jannetti, Eckert, Gaub, Chhabra and Pfaender199), and altered hemisphere-specific distribution, with a significant gain in the right hemisphere in pups (p = 0·017)(Reference Grabrucker, Haderspeck, Sauer, Kittelberger, Asoglu and Abaei173). Similarly, the PZD and homozygous Shank3B−/− mice showed both convergent and divergent brain region abnormalities(Reference Schoen, Asoglu, Bauer, Müller, Abaei and Sauer174). Prenatal Zn2+ is also crucial for gut formation in offspring(Reference Vela, Stark, Socha, Sauer, Hagmeyer and Grabrucker36). Indeed, Zn2+ deficiency causes the loss of brush border integrity and might lead to the dysfunction of disaccharidases, thereby altering gut maturation(Reference Wan and Zhang216). The offspring of an adult female swine fed a 250 ppm Zn2+ diet showed improved intestinal development(Reference Vela, Stark, Socha, Sauer, Hagmeyer and Grabrucker36). In addition to GI development, Zn2+ is an essential regulator of microbial composition and GI inflammation in ASD(Reference Sauer, Malijauskaite, Meleady, Boeckers, McGourty and Grabrucker175). Proteomic analysis of ileal samples from PZD mice revealed alterations in 183/1010 proteins involved in the pathways regulating the innate immune system, focal adhesions, TJ, and gap junctions(Reference Sauer, Malijauskaite, Meleady, Boeckers, McGourty and Grabrucker175). This suspected dysregulation of gut permeability was evidenced by increased serum Zonulin-1, a protein synthesized in the intestinal cells that regulates intestinal permeability in PZD. Gut inflammation was confirmed by a significant increase in serum inflammatory cytokines (IL1α, p = 0·037; IL1β, p = 0·050; IL7, p = 0·050; and IL17α, p = 0·011) in PZD pups(Reference Sauer, Malijauskaite, Meleady, Boeckers, McGourty and Grabrucker175) secondary to neuroinflammation(Reference Kirsten, Queiroz-Hazarbassanov, Bernardi and Felicio162). The microbiota of PZD mice showed a significant increase in Bacteriodetes and Actinobacteria (p < 0·001 and p < 0·001, respectively) and a significant decrease in Firmicutes (p < 0·001) at the phyla level, whereas at the genus level, PZD mice showed significantly reduced diversity (p < 0·05)(Reference Sauer, Malijauskaite, Meleady, Boeckers, McGourty and Grabrucker175). Learning and memory impairments have also been reported in rats(Reference Hagmeyer, Haderspeck and Grabrucker31,Reference Halas, Hunt and Eberhardt217,Reference Tahmasebi Boroujeni, Naghdi, Shahbazi, Farrokhi, Bagherzadeh and Kazemnejad218) . Concerning immune system regulation, diets containing < 1 mg/kg Zn2+/day appeared to cause thymus atrophy, thereby impacting the T-helper cells and having teratogenic effects, as evidenced by the resulting congenital defects in mouse offspring(Reference Sauer, Hagmeyer, Grabrucker, Holick and Nieves219,Reference Uriu-Adams and Keen220) . Such systemic inflammatory events and prenatal stress have been reported to aggravate Zn2+ deficiency in mothers, thereby increasing the risk of ASD in the offspring(Reference Sauer, Hagmeyer, Grabrucker, Holick and Nieves219,Reference Uriu-Adams and Keen220) . Although more controlled studies are warranted to prove the association between PZD and ASD, these studies emphasized the importance of sufficient Zn2+ availability, especially during the prenatal period, to ensure proper fetal neurodevelopment and possibly prevent nongenetic causes of ASD. Therefore, it is essential to understand whether Zn2+ supplementation can alleviate ASD-related symptoms or reduce the risk of ASD.

Effects of zinc supplementation

Grabrucker et al. advocated Zn2+ supplementation as a potential treatment for ASD, citing its essential roles in the neurological, GI, and immune systems, and its ability to reverse behavioral abnormalities observed in rodent models of ASD(Reference Indika, Frye, Rossignol, Owens, Senarathne and Grabrucker221). Overall, Zn2+ supplementation may be useful in young individuals with ASD, as summarized in Table 1. In particular, diarrhea in ASD-C has been linked to Zn2+ deficiency, as its supplementation significantly reduces this symptom by potentiating immune function and ameliorating neurosensory deficits associated with Zn2+ deficiency(Reference Vela, Stark, Socha, Sauer, Hagmeyer and Grabrucker36). As reviewed earlier, all the pathways altered by Zn2+ deficiency may contribute to the etiology and severity of ASD. Observing the actual effects of Zn2+ supplementation in a pediatric population with ASD, a supplementation trial of 12 weeks significantly improved ASD severity according to Childhood Autism Rating Scale (CARS) scores (p = 0·0002) in thirty children, which may be related to the regulation of the previously discussed pathways(Reference Meguid, Bjørklund, Gebril, Doşa, Anwar and Elsaeid125). Interestingly, there was also a significant improvement in gross motor development (TGMD-2). In addition, metallothionein and serum Zn2+ and Cu2+ levels were significantly ameliorated, thereby improving the Zn2+/Cu2+ ratio. An increase in metallothionein, following Zn2+ supplementation, was expected and has been previously documented(Reference Irato, Sturniolo, Giacon, Magro, D’Inca and Mestriner222). However, a reduction in the gene expression of metallothionine 1 (MT1), following dietary Zn2+ supplementation, was also observed, warranting further investigation on this association in ASD-C(Reference Meguid, Bjørklund, Gebril, Doşa, Anwar and Elsaeid125).

Table 1 Effect of Zn2+ supplementation on ASD-related features or genes expression in individuals with ASD and animal and cell culture models of ASD

↑: Increase; ↓: Decrease; AMPAR, α-amino-3-hydroxy-5-methyl-4isoxazolepropionic acid receptor; ASD, autism spectrum disorder; ASD-C, children with autism spectrum disorder; BDNF, brain derived neurotropic factor; BTBR, Black and Tan Brachyury; CARS, childhood autism rating scale; EGFP, enhanced green fluorescent protein; ppm, parts per million; KO, knockout; LPS, lipopolysaccharide; NMDAR, N-methyl-D-aspartate receptor; NS, not significant; PPR, AMPAR-mediated paired-pulse ratio; SNV, single nucleotide variation; WT, wild-type; Zn2+, zinc; ZnSO4, zinc sulfate.

Most studies of Zn2+ supplementation have been conducted using animal models. A six-week experimental trial on Shank3B−/− mice displaying ASD-like characteristics fed with a regular and Zn2+ supplemented diet found that Zn2+ supplementation prevented ASD-related repetitive and anxiety behaviors and deficits in social novelty recognition. These effects were attributed to the fact that the proteins encoded by the SHANK genes are regulated by Zn2+ ions, and increased Zn2+ levels are associated with improved scaffold protein formation and synaptic maturation(Reference Hagmeyer, Sauer and Grabrucker65,Reference Vyas, Lee, Jung and Montgomery223) . Zn2+ supplementation also increased the recruitment of Zn2+-sensitive SHANK2 to the synapses. Also, this protein possesses a SAM domain that can bind to Zn2+; therefore, it is possible that higher dietary Zn2+ intake increases SHANK2 recruitment as evidenced in Shank3B ex13–16−/− mice(Reference Fourie, Vyas, Lee, Jung, Garner and Montgomery210). SHANK2 is known for its role in the development of Zn2+-sensitive synapses and the control of postsynaptic density(Reference Vyas, Jung, Lee, Garner and Montgomery224). Notably, in vitro studies reported that Zn2+ supplementation was able to rescue and increase synaptic density in SHANK2 single nucleotide variation (SNV)-associated deficits and, in turn, their ability to promote synapse formation and maintenance(Reference Vyas, Jung, Lee, Garner and Montgomery224). Additional outcomes following Zn2+ supplementation include reduced synaptic transmission through the NMDA-type glutamate receptors and improvement of the slow decay of NMDAR-mediated currents(Reference Fourie, Vyas, Lee, Jung, Garner and Montgomery210). The Zn2+-mediated mitigation of ASD-like symptoms co-occurs with its modulation of NMDA current, suggesting that the NMDAR may serve as targets for Zn2+ dietary changes(Reference Fourie, Vyas, Lee, Jung, Garner and Montgomery210). Similarly, 150 ppm Zn2+ supplementation in another 9–11 week trial using the rodent model of ASD, Tbr1+/− (Tbr1 haploinsufficiency), prevented impairments in auditory fear memory and social interaction and significantly restored the synaptic puncta density of GluN1, which is essential for functional NMDAR as well as SHANK3 expression(Reference Lee, Jung, Vyas, Skelton, Abraham and Hsueh202). In LPS-exposed pregnant Wistar rats, ASD-like behavioral, brain, and immune disturbances were induced by significantly elevated levels of free-mature BDNF (p < 0·05). Supplementation of rats with Zn2+ reduced the elevation of LPS-induced BDNF to the same level as that in the control group(Reference Kirsten, Queiroz-Hazarbassanov, Bernardi and Felicio162). Furthermore, LPS exposure reduced the number of offspring, and treatment with Zn2+ prevented this reduction. Moreover, pups prenatally exposed to LPS spent significantly longer periods (p < 0·05) without calling their mothers, and posttreatment with Zn2+ prevented this LPS-induced behavioral impairment(Reference Kirsten, Queiroz-Hazarbassanov, Bernardi and Felicio162). This indicated that BDNF hyperactivity, as observed in individuals with ASD, may contribute to communication deficits that may be improved by Zn2+ supplementation. Moreover, microbial diversity seemed to be affected in those with ASD, and similar features were found in the Shank3B−/− mouse models, where significantly higher microbial diversity was observed in WT than mutated mice (p < 0·05)(Reference Wong, Jung, Lee, Fourie, Handley and Montgomery209). Overall, Muribaculaceae (Bacteroidetes) and Lachnospiraceae (Firmicutes) were significantly differentially represented between WT and Shank3B−/− mice fed Zn2+ (30 ppm) (p < 0·05). An experimental trial reported improved ASD-like characteristics in offspring born from Shank3B−/− mice with maternal Zn2+ supplementation and improved social interactions in the offspring(Reference Lee, Jung, Vyas, Skelton, Abraham and Hsueh202). It is important to note that the rescue effect of Zn2+ on social behaviors was maintained throughout adulthood in mice (week 16 of age), indicating the long-lasting benefits of Zn2+ supplementation during early brain development(Reference Vyas, Lee, Jung and Montgomery223). Furthermore, there were no significant differences in excessive grooming behavior in adult mice or anxiety behavior in juvenile and adult mice compared with the offspring born to WT mice(Reference Vyas, Lee, Jung and Montgomery223). In line with the three diagnostic features of ASD, BTBR T + tf/J (BTBR) mice displayed behaviors consistent with them, such as impaired social interaction and communication, as well as increased repetitive behaviors(Reference Meyza, Defensor, Jensen, Corley, Pearson and Pobbe225). A recent study compared BTBR mice with the C57BL/6 WT strain for several baseline ASD-like characteristics and then investigated the effect of Zn2+ supplementation using 60 ppm of Zn2+ in water(Reference Zhang, Xu, Ma, Wang, Jin and Li226). Specifically, Zn2+ supplemented water increased the social preference and social novelty indices in the BTBR mice, as determined by a three-chambered socialization experiment (p < 0·001), indicating improved social behavior. Zn2+-supplemented water also significantly improved repetitive behavior in the mice, as indicated by the marble burying and self-grooming tests. The BTBR mice had a significantly higher rate of marble burial than WT mice (p < 0·01) before treatment, which was significantly reduced in the BTBR mice (p < 0·05) post-treatment. In addition, the Holm–Sidak test confirmed that self-grooming time was significantly increased in the BTBR mice compared with that in the C57BL/6 mice (p < 0·001); whereas, self-grooming time was significantly reduced in BTBR mice post-Zn2+ treatment (p < 0·001). Finally, Zn2+-supplemented water reduced anxiety-like behavior, as confirmed by the open field test. Compared with WT mice, the BTBR mice displayed a more anxious behavior, which was significantly reduced post-Zn2+ water treatment (p < 0·05). Moreover, this treatment significantly reduced the convulsion susceptibility of the BTBR mice (p < 0·05) compared with that of the WT mice. It also boosted hippocampal cell growth, indicating that Zn2+ supplementation significantly restored neuronal proliferation in the hippocampus of the BTBR mice (p < 0·05). Collectively, these investigations suggested that Zn2+ can help mitigate autism-related symptoms in the BTBR model of autism(Reference Zhang, Xu, Ma, Wang, Jin and Li226) and these studies highlighted that Zn2+ may play a significant role from gestation throughout adulthood.

Concluding remarks

Current evidence indicates that Zn2+ dyshomeostasis, resulting from low levels of Zn2+ and reduced Zn2+/Cu2+ ratios, is a prevalent feature among individuals with ASD. PZD, low dietary Zn2+ intake, genetic makeup, and comorbidities associated with ASD may account for this condition.

Reduced Zn2+ availability, particularly during gestation, early life, and neurodevelopment, may alter Zn2+ homeostasis in the brain and zincergic innervation. Zn2+, being a potent neuromodulator, plays a pivotal role in proper synaptic transmission and neuronal function in the areas of the brain disrupted in ASD. Neuroinflammation and systemic inflammation reported in Zn2+ deficient states and ASD could also alter brain performance via the microbiome and gut–brain axis.

Several animal studies have demonstrated that Zn2+ deficiency is associated with changes in gut bacterial composition at the phylum, genus, and species levels, disrupts TJ complexes, and worsens phenotypes in rodent models of ASD. Solid evidence using SHANK rodent models of ASD has demonstrated the importance of altered crosstalk between genetic defects, Zn2+ dyshomeostasis, and gut dysbiosis in the pathophysiology of ASD. These findings suggested that Zn2+ is an essential micronutrient for the survival of commensal microbes in the GI tract and critically controls TJ complexes in the gut epithelium, promoting gut wall integrity and preventing leaky gut syndrome. Notably, the beneficial effects of Zn2+ supplementation have been shown to improve the phenotype of rodent models, suggesting that Zn2+ plays a crucial role in maintaining synaptic transmission, neuronal function, and gut health.

Regrettably, studies in human individuals concerning Zn2+ dyshomeostasis in ASD are limited and often inconclusive. Although some findings support the hypothesis that Zn2+ supplementation can ameliorate the severity of ASD symptoms and slow its progression, the evidence remains insufficient and requires further validation. Few studies have comprehensively characterized the effect of Zn2+ deficiency on the gut microbiota in ASD-C, and the potential therapeutic efficacy of Zn2+ on SHANK-associated pathologies in individuals with ASD remain underexplored. The high heterogeneity of autism complicates our understanding and generalizability of these findings, highlighting a critical gap in literature. Therefore, the limited documented benefits of Zn2+ on ASD symptoms require additional clinical trials to validate its potential therapeutic use.

To further ongoing research, some of the key questions that need to be addressed are as follows: How does Zn2+ supplementation specifically affect neurodevelopmental outcomes in ASD-C? What are the optimal doses and forms of Zn2+ for therapeutic application? How do genetic and environmental factors modulate the relationship between Zn2+ levels and symptoms of ASD? To what extent can Zn2+ supplementation restore the gut-wall integrity and improve GI symptoms in ASD-C? What are the long-term effects of Zn2+ supplementation?

The potential impact of Zn2+ on synaptic transmission, brain development, and gut function strongly suggests that this trace mineral is a contributing factor of ASD. However, clarifying the association between Zn2+ and ASD is difficult owing to the high heterogeneity of autism. Since the prevalence of ASD is increasing at an alarming rate worldwide, comprehensive and coordinated scientific efforts are urgently required to accelerate the transition from bench work to bedside treatment and ensure the best possible outcomes for individuals with ASD.

Acknowledgements

The authors would like to thank the English language editing services provided by the United Arab Emirates University. The authors acknowledge that the graphical abstract and Figs. 24 were created using BioRender (www.biorender.com).

Authorship

Asma Ahmadani: conceptualization, writing—original draft, and writing—review & editing. Monia Kittana: writing—original draft, and writing—review & editing. Farah Al-Marzooq: writing—original draft, and writing—review & editing. Sandeep Subramanya: writing—original draft. Maria Cristina D’Adamo: writing—original draft. Amita Attlee: conceptualization, writing—review & editing, supervision, and funding acquisition. Mauro Pessia: conceptualization, writing—review & editing, supervision, and funding acquisition.

Financial support

The study was supported by the ASPIRE grant (21M149-AARE20-260) and the START-UP grant (G00003323), which was funded by the Office of the Provost for Research and Graduate Studies, United Arab Emirates University.

Competing interests

The authors declare no conflicts of interest.

References

American Psychiatric Association (2013) Diagnostic and Statistical Manual of Mental Disorders (5th ed., pp. 5059). Arlington, VA: American Psychiatric Publishing.Google Scholar
Mubashir, S, Farrugia, M, Coretti, L, Pessia, M & D’Adamo, MC (2020) Autism spectrum disorder. Matla Med J 32(3), 5666.Google Scholar
Lord, C, Elsabbagh, M, Baird, G & Veenstra-Vanderweele, J (2018) Autism spectrum disorder. Lancet 392(10146), 508520.Google Scholar
Fombonne, E (2018) The rising prevalence of autism. J Child Psychol Psychiat 59, 717720.Google Scholar
Grabrucker, AM, ed. (2021) Autism Spectrum Disorders [Internet]. Brisbane, AU: Exon Publications; [cited 2025 Apr 3]. Available from: http://www.ncbi.nlm.nih.gov/books/NBK573612/ Google Scholar
Bhandari, R, Paliwal, JK & Kuhad, A (2020) Neuropsychopathology of autism spectrum disorder: complex interplay of genetic, epigenetic, and environmental factors. In Essa, MM & Qoronfleh, MW, editors. Personalized Food Intervention and Therapy for Autism Spectrum Disorder Management [Internet]. Cham: Springer International Publishing; [cited 2022 May 29]. pp 97141. https://doi.org/10.1007/978-3-030-30402-7_4 Google Scholar
Cheroni, C, Caporale, N & Testa, G (2020) Autism spectrum disorder at the crossroad between genes and environment: contributions, convergences, and interactions in ASD developmental pathophysiology. Mol Autism 11(1), 69.Google Scholar
Imamura, A, Morimoto, Y, Ono, S, Kurotaki, N, Kanegae, S, Yamamoto, N, et al. (2020) Genetic and environmental factors of schizophrenia and autism spectrum disorder: insights from twin studies. J Neural Transm 127(11), 15011515.Google Scholar
Masini, E, Loi, E, Vega-Benedetti, AF, Carta, M, Doneddu, G, Fadda, R, et al. (2020) An overview of the main genetic, epigenetic and environmental factors involved in autism spectrum disorder focusing on synaptic activity. Int J Mol Sci 21(21), 8290.Google Scholar
Saxena, R, Babadi, M, Namvarhaghighi, H & Roullet, FI (2020) Chapter two – role of environmental factors and epigenetics in autism spectrum disorders. In: Ilieva, M & Lau, WKW, editors. Progress in Molecular Biology and Translational Science [Internet]. Academic Press; [cited 2022 May 29]. pp 3560. (Autism; vol. 173). Available from: https://www.sciencedirect.com/science/article/pii/S1877117320300697 Google Scholar
Yoon, SH, Choi, J, Lee, WJ & Do, JT (2020) Genetic and epigenetic etiology underlying autism spectrum disorder. J Clin Med 9(4), 966.Google Scholar
Alsufiani, HM, Alkhanbashi, AS, Laswad, NAB, Bakhadher, KK, Alghamdi, SA, Tayeb, HO, et al. (2022) Zinc deficiency and supplementation in autism spectrum disorder and Phelan–McDermid syndrome. J Neurosci Res 100(4), 970978.Google Scholar
Babaknejad, N, Sayehmiri, F, Sayehmiri, K, Mohamadkhani, A & Bahrami, S (2016) The relationship between zinc levels and autism: a systematic review and meta-analysis. Iran J Child Neurol 10(4), 19.Google Scholar
Maret, W & Sandstead, HH (2006) Zinc requirements and the risks and benefits of zinc supplementation. J Trace Elem Med Biol 20(1), 318.Google Scholar
Hara, T, Takeda, T, Takagishi, T, Fukue, K, Kambe, T & Fukada, T (2017) Physiological roles of zinc transporters: molecular and genetic importance in zinc homeostasis. J Physiol Sci 67(2), 283301.Google Scholar
Kambe, T, Tsuji, T, Hashimoto, A & Itsumura, N (2015) The physiological, biochemical, and molecular roles of zinc transporters in zinc homeostasis and metabolism. Physiol Rev 95(3), 749784.Google Scholar
Fukada, T, Yamasaki, S, Nishida, K, Murakami, M, Hirano, T (2011) Zinc homeostasis and signaling in health and diseases: zinc signaling. J Biol Inorg Chem 16(7), 11231134.Google Scholar
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(11), e50568.Google Scholar
Kumera, G, Awoke, T, Melese, T, Eshetie, S, Mekuria, G, Mekonnen, F, et al. (2015) Prevalence of zinc deficiency and its association with dietary, serum albumin and intestinal parasitic infection among pregnant women attending antenatal care at the University of Gondar Hospital, Gondar, Northwest Ethiopia. BMC Nutr 1(1), 31.Google Scholar
Prasad, AS (2017) Discovery of zinc for human health and biomarkers of zinc deficiency. In Collins, J. F. (Ed.), Molecular, Genetic, and Nutritional Aspects of Major and Trace Minerals (pp. 241260). London: Academic Press. https://doi.org/10.1016/B978-0-12-802168-2.00020-8.Google Scholar
De Benedictis, CA, Trame, S, Rink, L & Grabrucker, AM (2023) Prevalence of low dietary zinc intake in women and pregnant women in Ireland. Ir J Med Sci 192(4), 18351845.Google Scholar
Scarpellini, E, Balsiger, LM, Maurizi, V, Rinninella, E, Gasbarrini, A, Giostra, N, et al. (2022) Zinc and gut microbiota in health and gastrointestinal disease under the COVID-19 suggestion. BioFactors 48(2), 294306.Google Scholar
Zhong, W, Yang, C, Zhu, L, Huang, YQ & Chen, YF (2020) Analysis of the relationship between the mutation site of the SLC39A4 gene and acrodermatitis enteropathica by reporting a rare Chinese twin: a case report and review of the literature. BMC Pediatr 20(1), 34.Google Scholar
McClain, C, Vatsalya, V & Cave, M (2017) Role of zinc in the development/progression of alcoholic liver disease. Curr Treat Options Gastroenterol 15(2), 285295.Google Scholar
Corbo, MD & Lam, J (2013) Zinc deficiency and its management in the pediatric population: a literature review and proposed etiologic classification. J Am Acad Dermatol 69(4), 616624.e1.Google Scholar
Kambe, T, Fukue, K, Ishida, R & Miyazaki, S (2015) Overview of inherited zinc deficiency in infants and children. J Nutr Sci Vitaminol 61(Supplement), S44S46.Google Scholar
Van Wouwe, JP (1995) Clinical and laboratory assessment of zinc deficiency in Dutch children. Biol Trace Elem Res 49(2), 211225.Google Scholar
Willoughby, JL & Bowen, CN (2014) Zinc deficiency and toxicity in pediatric practice. Curr Opin Pediatr 26(5), 579584.Google Scholar
Grønli, O, Kvamme, JM, Friborg, O & Wynn, R (2013) Zinc deficiency is common in several psychiatric disorders. PloS One 8(12), e82793.Google Scholar
El-Bakry (2023) Zinc deficiency in children with attention-deficit hyperactivity disorder [Internet]. [cited 2023 Jan 20]. Available from: https://new.ejpsy.eg.net/article.asp?issn=1110-1105;year=2019;volume=40;issue=2;spage=95;epage=103;aulast=El-Bakry Google Scholar
Hagmeyer, S, Haderspeck, JC, Grabrucker, AM (2015) Behavioral impairments in animal models for zinc deficiency. Front Behav Neurosci 8, 443.Google Scholar
Sweetman, DU, O’Donnell, SM, Lalor, A, Grant, T, Greaney, H (2019) Zinc and vitamin A deficiency in a cohort of children with autism spectrum disorder. Child: Care, Health Dev 45(3), 380386.Google Scholar
Cherif, L, Boudabous, J, Khemekhem, K, Mkawer, S, Ayadi, H & Moalla, Y (2018) Feeding problems in children with autism spectrum disorders. J Fam Med 1(1), 3039.Google Scholar
Sharp, WG, Berry, RC, McCracken, C, Nuhu, NN, Marvel, E, Saulnier, CA, et al. (2013) Feeding problems and nutrient intake in children with autism spectrum disorders: a meta-analysis and comprehensive review of the literature. J Autism Dev Disord 43(9), 21592173.Google Scholar
Bjørklund, G, Meguid, NA, El-Bana, MA, Tinkov, AA, Saad, K, Dadar, M, et al. (2020) Oxidative stress in autism spectrum disorder. Mol Neurobiol 57(5), 23142332.Google Scholar
Vela, G, Stark, P, Socha, M, Sauer, AK, Hagmeyer, S & Grabrucker, AM (2015) Zinc in gut-brain interaction in autism and neurological disorders. Neural Plast 2015. Article ID 972791. https://doi.org/10.1155/2015/972791.Google Scholar
Talantseva, OI, Romanova, RS, Shurdova, EM, Dolgorukova, TA, Sologub, PS, Titova, OS, et al. (2023) The global prevalence of autism spectrum disorder: a three-level meta-analysis. Front Psychiatry [Internet]. [cited 2024 May 7]; 14. Available from: https://www.frontiersin.org/journals/psychiatry/articles/10.3389/fpsyt.2023.1071181/full Google Scholar
Fombonne, E, MacFarlane, H & Salem, AC (2021) Epidemiological surveys of ASD: advances and remaining challenges. J Autism Dev Disorders 51(12), 42714290.Google Scholar
Maenner, MJ (2020) Prevalence and characteristics of autism spectrum disorder among children aged 8 years — autism and developmental disabilities monitoring network, 11 sites, United States. MMWR Surveill Summ [Internet]. [cited 2024 May 30]; 72. Available from: https://www.cdc.gov/mmwr/volumes/72/ss/ss7202a1.htm Google Scholar
Canada PHA of (2022) Autism spectrum disorder: highlights from the 2019 Canadian health survey on children and youth [Internet]. [cited 2024 May 30]. Available from: https://www.canada.ca/en/public-health/services/publications/diseases-conditions/autism-spectrum-disorder-canadian-health-survey-children-youth-2019.html Google Scholar
Linnsand, P, Gillberg, C, Nilses, Å, Hagberg, B & Nygren, G (2021) A high prevalence of autism spectrum disorder in preschool children in an immigrant, multiethnic population in Sweden: challenges for health care. J Autism Dev Disord 51(2), 538549.Google Scholar
O’Nions, E, Petersen, I, Buckman, JEJ, Charlton, R, Cooper, C, Corbett, A, et al. (2023) Autism in England: assessing underdiagnosis in a population-based cohort study of prospectively collected primary care data. Lancet Regional Health – Europe [Internet]. [cited 2024 May 30]; 29. Available from: https://www.thelancet.com/journals/lanepe/article/PIIS2666-7762(23)00045-5/fulltext#secsectitle0120 Google Scholar
Zeidan, J, Fombonne, E, Scorah, J, Ibrahim, A, Durkin, MS, Saxena, S, et al. (2022) Global prevalence of autism: a systematic review update. Autism Res 15(5), 778790.Google Scholar
Saito, M, Hirota, T, Sakamoto, Y, Adachi, M, Takahashi, M, Osato-Kaneda, A, et al. (2020) Prevalence and cumulative incidence of autism spectrum disorders and the patterns of co-occurring neurodevelopmental disorders in a total population sample of 5-year-old children. Mol Autism 11(1), 35.Google Scholar
Sung, M, Magiati, I, Goh, T, Fung, D, Aljunied, M, Phua, D, et al. (2020) Singapore and Autism Spectrum Disorder. Encyclopedia Springer [Internet]. [cited 2024 May 30]; Available from: https://www.academia.edu/download/110133440/978-1-4614-6435-8_102038-2.pdf. https://doi.org/10.1007/978-1-4614-6435-8_102038-2Google Scholar
Tiwari, R, Purkayastha, K & Gulati, S (2021) Public health dimensions of autism spectrum disorder in India: an overview. J Compr Health 9(2), 5762.Google Scholar
Rasool, A, Rasool, SH, Siddique, M, Bajwa, N, Khan, RM, Rasool, A, et al. (2023) Prevalence of autism spectrum disorders in Asian countries. J Soc Prev Advoc Res KEMU 2(3), 215224.Google Scholar
May, T, Brignell, A & Williams, K (2020) Autism spectrum disorder prevalence in children aged 12–13 years from the longitudinal study of Australian children. Autism Res 13(5), 821827.Google Scholar
Bowden, N, Thabrew, H, Kokaua, J, Audas, R, Milne, B, Smiler, K, et al. (2020) Autism spectrum disorder/Takiwātanga: an integrated data infrastructure-based approach to autism spectrum disorder research in New Zealand. Autism 24(8), 22132227.Google Scholar
Lagunju, IA, Bella-Awusah, TT & Omigbodun, OO (2014) Autistic disorder in Nigeria: profile and challenges to management. Epilepsy Behav 39, 126129.Google Scholar
Pillay, S, Duncan, M & de Vries, PJ (2021) Autism in the Western Cape province of South Africa: rates, socio-demographics, disability and educational characteristics in one million school children. Autism 25(4), 10761089.Google Scholar
Virolainen, S, Hussien, W & Dalibalta, S (2020) Autism spectrum disorder in the United Arab Emirates: potential environmental links. Rev Environ Health 35(4), 359369.Google Scholar
Alshaban, F, Aldosari, M, Al-Shammari, H, El-Hag, S, Ghazal, I, Tolefat, M, et al. (2019) Prevalence and correlates of autism spectrum disorder in Qatar: a national study. J Child Psychol Psychiatry 60(12), 12541268.Google Scholar
Sabbagh, HJ, Al-Jabri, BA, Alsulami, MA, Hashem, LA, Aljubour, AA & Alamoudi, RA (2021) Prevalence and characteristics of autistic children attending autism centres in 2 major cities in Saudi Arabia. Saudi Med J 42(4), 419427.Google Scholar
Salhia, HO, Al-Nasser, LA, Taher, LS, Al-Khathaami, AM & El-Metwally, AA (2014) Systemic review of the epidemiology of autism in Arab Gulf countries. Neurosciences (Riyadh) 19(4), 291296.Google Scholar
Al-Mamri, W, Idris, AB, Dakak, S, Al-Shekaili, M, Al-Harthi, Z, Alnaamani, AM, et al. (2019) Revisiting the prevalence of autism spectrum disorder among Omani children. Sultan Qaboos Univ Med J 19(4), e305e309.Google Scholar
Mottron, L, Duret, P, Mueller, S, Moore, RD, Forgeot d’Arc, B, Jacquemont, S, et al. (2015) Sex differences in brain plasticity: a new hypothesis for sex ratio bias in autism. Mol Autism 6(1), 33.Google Scholar
Daghsni, M, Rima, M, Fajloun, Z, Ronjat, M, Brusés, JL, M’rad, R, et al. (2018) Autism throughout genetics: perusal of the implication of ion channels. Brain Behav 8(8), e00978.Google Scholar
Zoghbi, HY & Bear, MF (2012) Synaptic dysfunction in neurodevelopmental disorders associated with autism and intellectual disabilities. Cold Spring Harb Perspect Biol 4(3), a009886.Google Scholar
Lyall, K, Croen, L, Daniels, J, Fallin, MD, Ladd-Acosta, C, Lee, BK, et al. (2017) The changing epidemiology of autism spectrum disorders. Annu Rev Public Health 38(1), 81102.Google Scholar
Sandin, S, Lichtenstein, P, Kuja-Halkola, R, Hultman, C, Larsson, H & Reichenberg, A (2017) The heritability of autism spectrum disorder. JAMA 318(12), 11821184.Google Scholar
Yousuf, F, Asif, AS, Husain, S & Rafiq, R (2023) Prevalence of autism spectrum disorder (ASD) in offspring’s of consanguineous marriages. J BioMed Res Rep 2(2). https://doi.org/10.59657/2837-4681.brs.23.014 Google Scholar
Yu, TW, Chahrour, MH, Coulter, ME, Jiralerspong, S, Okamura-Ikeda, K, Ataman, B, et al. (2013) Using whole-exome sequencing to identify inherited causes of autism. Neuron 77(2), 259273.Google Scholar
Huguet, G & Bourgeron, T (2016) Chapter 2 – genetic causes of autism spectrum disorders. In: Sala, C & Verpelli, C, editors. Neuronal and Synaptic Dysfunction in Autism Spectrum Disorder and Intellectual Disability [Internet]. San Diego: Academic Press; [cited 2022 Jan 29]. pp 1324. Available from: https://www.sciencedirect.com/science/article/pii/B9780128001097000029 Google Scholar
Hagmeyer, S, Sauer, AK & Grabrucker, AM (2018) Prospects of zinc supplementation in autism spectrum disorders and Shankopathies such as Phelan McDermid Syndrome. Front Synapt Neurosci 10, 11.Google Scholar
Muhle, R, Trentacoste, SV & Rapin, I (2004) The genetics of autism. Pediatrics 113(5), e472e486.Google Scholar
Ambrosini, E, Sicca, F, Brignone, MS, D’Adamo, MC, Napolitano, C, Servettini, I, et al. (2014) Genetically induced dysfunctions of Kir2.1 channels: implications for short QT3 syndrome and autism–epilepsy phenotype. Hum Mol Genet 23(18), 48754886.Google Scholar
D’Adamo, MC, Catacuzzeno, L, Di Giovanni, G, Franciolini, F & Pessia, M (2013) K+ channelepsy: progress in the neurobiology of potassium channels and epilepsy. Front Cell Neurosci 7, 134.Google Scholar
Guglielmi, L, Servettini, I, Caramia, M, Catacuzzeno, L, Franciolini, F, D’Adamo, MC, et al. (2015) Update on the implication of potassium channels in autism: K+ channel autism spectrum disorder. Front Cell Neurosci 9, 34.Google Scholar
Kim, JB (2014) Channelopathies. Korean J Pediatr 57(1), 118.Google Scholar
Sicca, F, Imbrici, P, D’Adamo, MC, Moro, F, Bonatti, F, Brovedani, P, et al. (2011) Autism with seizures and intellectual disability: possible causative role of gain-of-function of the inwardly-rectifying K+ channel Kir4. 1. Neurobiol Dis 43(1), 239247.Google Scholar
Sicca, F, Ambrosini, E, Marchese, M, Sforna, L, Servettini, I, Valvo, G, et al. (2016) Gain-of-function defects of astrocytic Kir4.1 channels in children with autism spectrum disorders and epilepsy. Sci Rep 6(1), 34325.Google Scholar
Peralta, FA, Huidobro-Toro, JP (2016) Zinc as allosteric ion channel modulator: ionotropic receptors as metalloproteins. Int J Mol Sci 17(7), 1059.Google Scholar
Noh, S, Lee, SR, Jeong, YJ, Ko, KS, Rhee, BD, Kim, N, et al. (2015) The direct modulatory activity of zinc toward ion channels. Integr Med Res 4(3), 142146.Google Scholar
D’Adamo, MC, Moro, F, Imbrici, P, Martino, D, Roscini, M, Santorelli, FM, et al. (2011) The emerging role of the inwardly rectifying K+ channels in autism spectrum disorders and epilepsy. Malta Medical Journal, 23(3), 1014.Google Scholar
Rylaarsdam, L & Guemez-Gamboa, A (2019) Genetic causes and modifiers of autism spectrum disorder. Front Cell Neurosci [Internet]. [cited 2022 Jan 29]; 13. Available from: https://www.frontiersin.org/article/10.3389/fncel.2019.00385 Google Scholar
SFARI [Internet]. [cited 2025 Jan 15]. Simons Foundation Autism Research Initiative. Available from: https://www.sfari.org/ Google Scholar
Bölte, S, Girdler, S & Marschik, PB (2019) The contribution of environmental exposure to the etiology of autism spectrum disorder. Cell Mol Life Sci 76(7), 1275.Google Scholar
Ratajczak, HV (2011) Theoretical aspects of autism: causes—a review. J Immunotoxicol 8(1), 6879.Google Scholar
Alkhalidy, H, Abushaikha, A, Alnaser, K, Obeidat, MD & Al-Shami, I (2021) Nutritional status of pre-school children and determinant factors of autism: a case–control study. Front Nutr [Internet]. [cited 2022 Feb 8]; 8. Available from: https://www.frontiersin.org/article/10.3389/fnut.2021.627011 Google Scholar
Cortés-Albornoz, MC, García-Guáqueta, DP, Velez-van-Meerbeke, A & Talero-Gutiérrez, C (2021) Maternal nutrition and neurodevelopment: a scoping review. Nutrients 13(10), 3530.Google Scholar
Vecchione, R, Wang, S, Rando, J, Chavarro, JE, Croen, LA, Fallin, MD, et al. (2022) Maternal dietary patterns during pregnancy and child autism-related traits: results from two US cohorts. Nutrients 14(13), 2729.Google Scholar
Veniaminova, E, Cespuglio, R, Cheung, CW, Umriukhin, A, Markova, N, Shevtsova, E, et al. (2017) Autism-like behaviours and memory deficits result from a Western diet in mice. Neural Plast 2017(1), 9498247.Google Scholar
Gogou, M & Kolios, G (2020) Nutritional supplements during gestation and autism spectrum disorder: what do we really know and how far have we gone? J Am Coll Nutr 39(3), 261271.Google Scholar
Bragg, M, Chavarro, JE, Hamra, GB, Hart, JE, Tabb, LP, Weisskopf, MG, et al. (2022) Prenatal diet as a modifier of environmental risk factors for autism and related neurodevelopmental outcomes. Curr Envir Health Rep 9(2), 324338.Google Scholar
Akdas, S & Yazihan, N (2020) Cord blood zinc status effects on pregnancy outcomes and its relation with maternal serum zinc levels: a systematic review and meta-analysis. World J Pediatr 16(4), 366376.Google Scholar
Grzeszczak, K, Kwiatkowski, S & Kosik-Bogacka, D (2020) The role of Fe, Zn, and Cu in pregnancy. Biomolecules 10(8), 1176.Google Scholar
Iqbal, S & Ali, I (2021) Effect of maternal zinc supplementation or zinc status on pregnancy complications and perinatal outcomes: an umbrella review of meta-analyses. Heliyon 7(7), e07540.Google Scholar
Saghazadeh, A, Ahangari, N, Hendi, K, Saleh, F & Rezaei, N (2017) Status of essential elements in autism spectrum disorder: systematic review and meta-analysis. Rev Neurosci 28(7), 783809.Google Scholar
Faber, S, Zinn, GM, Kern, JC II & Skip Kingston, HM (2009) The plasma zinc/serum copper ratio as a biomarker in children with autism spectrum disorders. Biomarkers 14(3), 171180.Google Scholar
Alves, CX, de Brito, NJN, Vermeulen, KM, Dantas Lopes, MMG, França, MC, Bruno, SS, et al. (2016) Serum zinc reference intervals and its relationship with dietary, functional, and biochemical indicators in 6- to 9-year-old healthy children. Food Nutr Res 60. https://doi.org/10.3402/fnr.v60.30157 Google Scholar
Li, S, Wang, JL, Bjørklund, G, Zhao, WN & Yin, C (2014) Serum copper and zinc levels in individuals with autism spectrum disorders. NeuroReport 25(15), 12161220.Google Scholar
Crăciun, EC, Bjørklund, G, Tinkov, AA, Urbina, MA, Skalny, AV, Rad, F, et al. (2016) Evaluation of whole blood zinc and copper levels in children with autism spectrum disorder. Metab Brain Dis 31(4), 887890.Google Scholar
do Nascimento, PK da SB, Oliveira Silva, DF, de Morais, TLSA, de Rezende, AA (2023) Zinc status and autism spectrum disorder in children and adolescents: a systematic review. Nutrients 15(16), 3663.Google Scholar
Guo, M, Li, L, Zhang, Q, Chen, L, Dai, Y, Liu, L, et al. (2020) Vitamin and mineral status of children with autism spectrum disorder in Hainan Province of China: associations with symptoms. Nutr Neurosci 23(10), 803810.Google Scholar
Wu, J, Wang, D, Yan, L, Jia, M, Zhang, J, Han, S, et al. (2022) Associations of essential element serum concentrations with autism spectrum disorder. Environ Sci Pollut Res [Internet]. [cited 2022 Aug 2]. https://doi.org/10.1007/s11356-022-21978-1 Google Scholar
Feng, J, Shan, L, Miao, C, Xue, Y, Yue, X & Jia, F (2023) The association of vitamin A, zinc and copper levels with clinical symptoms in children with autism spectrum disorders in Jilin Province, China. BMC Pediatr 23(1), 173.Google Scholar
Siddiqi, UR, Begum, S, Shahjadi, S, Afroz, S, Mahruba, SN, Parvin, J, et al. (2023) Plasma zinc, copper and serum ceruloplasmin levels of autism spectrum disorder children in Bangladesh. Heliyon [Internet]. [cited 2024 May 6]; 9(8). Available from: https://www.cell.com/heliyon/pdf/S2405-8440(23)05832-2.pdf Google Scholar
Saldanha Tschinkel, PF, Bjørklund, G, Conón, LZZ, Chirumbolo, S, Nascimento, VA (2018) Plasma concentrations of the trace elements copper, zinc and selenium in Brazilian children with autism spectrum disorder. Biomed Pharmacother 106, 605609.Google Scholar
Hawari, I, Eskandar, MB & Alzeer, S (2020) The role of lead, manganese, and zinc in autism spectrum disorders (ASDs) and attention-deficient hyperactivity disorder (ADHD): a case-control study on Syrian children affected by the Syrian crisis. Biol Trace Elem Res 197(1), 107114.Google Scholar
Zinc – Livingstone (2015) Nutrition in Clinical Practice. Wiley Online Library [Internet]. [cited 2021 Dec 29]. Available from: https://aspenjournals.onlinelibrary.wiley.com/doi/full/10.1177/0884533615570376 Google Scholar
Wieringa, FT, Dijkhuizen, MA, Fiorentino, M, Laillou, A & Berger, J (2015) Determination of zinc status in humans: which indicator should we use? Nutrients 7(5), 32523263.Google Scholar
Roohani, N, Hurrell, R, Kelishadi, R & Schulin, R (2013) Zinc and its importance for human health: an integrative review. J Res Med Sci 18(2), 144157.Google Scholar
Lowe, NM, Fekete, K & Decsi, T (2009) Methods of assessment of zinc status in humans: a systematic review. Am J Clin Nutr 89(6), 2040S2051S.Google Scholar
DiBaise, M & Tarleton, SM (2019) Hair, nails, and skin: differentiating cutaneous manifestations of micronutrient deficiency. Nutr Clin Pract 34(4), 490503.Google Scholar
Fiore, M, Barone, R, Copat, C, Grasso, A, Cristaldi, A, Rizzo, R, et al. (2020) Metal and essential element levels in hair and association with autism severity. J Trace Elem Med Biol 57, 126409.Google Scholar
Blaurock-Busch, E, Amin, OR, Dessoki, HH & Rabah, T (2012) Toxic metals and essential elements in hair and severity of symptoms among children with autism. Maedica (Bucur) 7(1), 3848.Google Scholar
Yasuda, H, Yoshida, K, Yasuda, Y & Tsutsui, T (2011) Infantile zinc deficiency: association with autism spectrum disorders. Sci Rep 1(1), 129.Google Scholar
Kaczmarek, KA, Dobrzyńska, M & Drzymała-Czyż, S (2024) Iron, magnesium, zinc and selenium – the most common elemental deficiencies in children with autism spectrum disorder. Res Autism Spectr Disorders 110, 102288.Google Scholar
Bjørklund, G (2013) The role of zinc and copper in autism spectrum disorders. Acta Neurobiol Exp (Wars) 73(2), 225236.Google Scholar
Baj, J, Flieger, W, Flieger, M, Forma, A, Sitarz, E, Skórzyńska-Dziduszko, K, et al. (2021) Autism spectrum disorder: trace elements imbalances and the pathogenesis and severity of autistic symptoms. Neurosci Biobehav Rev 129, 117132.Google Scholar
Ramaekers, VT, Sequeira, JM, Thöny, B & Quadros, EV (2020) Oxidative stress, folate receptor autoimmunity, and CSF findings in severe infantile autism. Autism Res Treat 2020. Article ID 9095284. https://doi.org/10.1155/2020/90952 Google Scholar
Gropper, S, Smith, J & Carr, T (2022) Advanced Nutrition and Human Metabolism. 8th ed. Boston, MA: Cengage Learning.Google Scholar
To, P, Do, M, Cho, JH & Jung, C (2020) Growth modulatory role of zinc in prostate cancer and application to cancer therapeutics. Int J Mol Sci 21, 2991.Google Scholar
Ross, MM, Hernandez-Espinosa, DR & Aizenman, E (2023) Neurodevelopmental consequences of dietary zinc deficiency: a status report. Biol Trace Elem Res 201(12), 56165639.Google Scholar
Marger, L, Schubert, CR & Bertrand, D (2014) Zinc: an underappreciated modulatory factor of brain function. Biochem Pharmacol 91(4), 426435.Google Scholar
DeBenedictis, CA, Raab, A, Ducie, E, Howley, S, Feldmann, J & Grabrucker, AM (2020) Concentrations of essential trace metals in the brain of animal species—a comparative study. Brain Sci 10(7), 460.Google Scholar
Gower-Winter, SD & Levenson, CW (2012) Zinc in the central nervous system: from molecules to behavior. BioFactors 38(3), 186193.Google Scholar
National Institute of Health. Office of dietary supplements. Zinc [Internet]. 2021 [cited 2022 Feb 2]. Available from: https://ods.od.nih.gov/factsheets/Zinc-HealthProfessional/ Google Scholar
Lönnerdal, B (2000) Dietary factors influencing zinc absorption. J Nutr 130(5), 1378S1383S.Google Scholar
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.Google Scholar
Jeong, J & Eide, DJ (2013) The SLC39 family of zinc transporters. Mol Aspects Med 34(2), 612619.Google Scholar
Hashimoto, A & Kambe, T (2022) Overview of the zinc absorption mechanism for improving zinc nutrition. Metallom Res 2(1), rev-20.Google Scholar
Martin, AB, Aydemir, TB, Guthrie, GJ, Samuelson, DA, Chang, SM & Cousins, RJ (2013) Gastric and colonic zinc transporter ZIP11 (Slc39a11) in mice responds to dietary zinc and exhibits nuclear localization123. J Nutr 143(12), 18821888.Google Scholar
Meguid, NA, Bjørklund, G, Gebril, OH, Doşa, MD, Anwar, M, Elsaeid, A, et al. (2019) The role of zinc supplementation on the metallothionein system in children with autism spectrum disorder. Acta Neurol Belg 119(4), 577583.Google Scholar
Cousins, RJ (2010) Gastrointestinal factors influencing zinc absorption and homeostasis. Int J Vitam Nutr Res 80(45), 243248.Google Scholar
Milne, DB, Canfield, WK, Mahalko, JR & Sandstead, HH (1983) Effect of dietary zinc on whole body surface loss of zinc: impact on estimation of zinc retention by balance method. Am J Clin Nutr 38(2), 181186.Google Scholar
Solomons, NW (2003) ZINC | physiology. In: Caballero, B, editor. Encyclopedia of Food Sciences and Nutrition. 2nd Edition [Internet]. Oxford: Academic Press; [cited 2022 Jun 3]. pp 62726277. Available from: https://www.sciencedirect.com/science/article/pii/B012227055X013092 Google Scholar
Sun, Q, Li, Q, Zhong, W, Zhang, J, Sun, X, Tan, X, et al. (2014) Dysregulation of hepatic zinc transporters in a mouse model of alcoholic liver disease. Am J Physiol Gastrointest Liver Physiol 307(3), G313G322.Google Scholar
Gerber, PA, Bellomo, EA, Hodson, DJ, Meur, G, Solomou, A, Mitchell, RK, et al. (2014) Hypoxia lowers SLC30A8/ZnT8 expression and free cytosolic Zn2+ in pancreatic beta cells. Diabetologia 57(8), 16351644.Google Scholar
Baltaci, AK & Mogulkoc, R (2007) Pinealectomy and melatonin administration in rats: their effects on plasma leptin levels and relationship with zinc. Biol Futur 58(4), 335343.Google Scholar
Unal, O, Baltaci, AK, Mogulkoc, R & Avunduk, MC (2021) Effect of pinealectomy and melatonin supplementation on metallothionein, ZnT2, ZIP2, ZIP4 and zinc levels in rat small intestine. Biotechn Histochem 96(8), 623635.Google Scholar
Hennigar, SR & McClung, JP (2016) Hepcidin attenuates zinc efflux in Caco-2 cells. J Nutr 146(11), 21672173.Google Scholar
Lee, JY, Kim, JH, Hong, SH, Lee, JY, Cherny, RA, Bush, AI, et al. (2004) Estrogen decreases zinc transporter 3 expression and synaptic vesicle zinc levels in mouse brain. J Biol Chem 279(10), 86028607.Google Scholar
Egefjord, L, Jensen, JL, Bang-Berthelsen, CH, Petersen, AB, Smidt, K, Schmitz, O, et al. (2009) Zinc transporter gene expression is regulated by pro-inflammatory cytokines: a potential role for zinc transporters in beta-cell apoptosis? BMC Endocr Disorders 9(1), 7.Google Scholar
Muayed, ME, Billings, LK, Raja, MR, Zhang, X, Park, PJ, Newman, MV, et al. (2010) Acute cytokine-mediated downregulation of the zinc transporter ZnT8 alters pancreatic β-cell function. J Endocrinol 206(2), 159169.Google Scholar
Gammoh, NZ & Rink, L (2017) Zinc in infection and inflammation. Nutrients 9(6), 624.Google Scholar
King, JC (2018) Yet again, serum zinc concentrations are unrelated to zinc intakes. J Nutr 148(9), 13991401.Google Scholar
Hashimoto, A, Nakagawa, M, Tsujimura, N, Miyazaki, S, Kizu, K, Goto, T, et al. (2016) Properties of Zip4 accumulation during zinc deficiency and its usefulness to evaluate zinc status: a study of the effects of zinc deficiency during lactation. Am J Physiol Regul Integr Comp Physiol 310(5), R459R468.Google Scholar
Wani, AL, Ansari, MO, Ahmad, MF, Parveen, N, Siddique, HR, Shadab, GGHA (2019) Influence of zinc levels on the toxic manifestations of lead exposure among the occupationally exposed workers. Environ Sci Pollut Res Int 26(32), 3354133554.Google Scholar
Bushnell, PJ & Levin, ED (1983) Effects of zinc deficiency on lead toxicity in rats. Neurobehav Toxicol Teratol 5(3), 283288.Google Scholar
Vance, TM & Chun, OK (2015) Zinc intake is associated with lower cadmium burden in U.S. adults. J Nutr 145(12), 27412748.Google Scholar
Porcheron, G, Garenaux, A, Proulx, J, Sabri, M & Dozois, C (2013) Iron, copper, zinc, and manganese transport and regulation in pathogenic Enterobacteria: correlations between strains, site of infection and the relative importance of the different metal transport systems for virulence. Front Cell Infect Microbiol [Internet]. [cited 2022 Jun 14]; 3. Available from: https://www.frontiersin.org/article/10.3389/fcimb.2013.00090 Google Scholar
Gabbianelli, R, Scotti, R, Ammendola, S, Petrarca, P, Nicolini, L, Battistoni, A (2011) Role of ZnuABC and ZinT in Escherichia coliO157:H7 zinc acquisition and interaction with epithelial cells. BMC Microbiol 11(1), 36.Google Scholar
Nielubowicz, GR, Smith, SN & Mobley, HLT (2010) Zinc uptake contributes to motility and provides a competitive advantage to Proteus mirabilis during experimental urinary tract infection. Infect Immun 78(6), 28232833.Google Scholar
Bersch, B, Bougault, C, Roux, L, Favier, A, Vernet, T & Durmort, C (2013) New Insights into Histidine Triad proteins: solution structure of a streptococcus pneumoniae PhtD domain and zinc transfer to AdcAII. PLoS One 8(11), e81168.Google Scholar
Liu, JZ, Jellbauer, S, Poe, A, Ton, V, Pesciaroli, M, Kehl-Fie, T, et al. (2012) Zinc sequestration by the neutrophil protein calprotectin enhances Salmonella growth in the inflamed gut. Cell Host Microbe 11(3), 227239.Google Scholar
Shoubridge, AP, Choo, JM, Martin, AM, Keating, DJ, Wong, ML, Licinio, J, et al. (2022) The gut microbiome and mental health: advances in research and emerging priorities. Mol Psychiatry 27(4), 19081919.Google Scholar
McDevitt, CA, Ogunniyi, AD, Valkov, E, Lawrence, MC, Kobe, B, McEwan, AG, et al. (2011) A molecular mechanism for bacterial susceptibility to zinc. PLOS Pathogens 7(11), e1002357.Google Scholar
Skalny, AV, Aschner, M, Lei, XG, Gritsenko, VA, Santamaria, A, Alekseenko, SI, et al. (2021) Gut microbiota as a mediator of essential and toxic effects of zinc in the intestines and other tissues. Int J Mol Sci 22(23), 13074.Google Scholar
Chen, L, Wang, Z, Wang, P, Yu, X, Ding, H, Wang, Z, et al. (2021) Effect of long-term and short-term imbalanced Zn manipulation on gut microbiota and screening for microbial markers sensitive to zinc status. Microbiol Spectr [Internet]. [cited 2022 Feb 17]; Available from: https://journals.asm.org/doi/abs/10.1128/Spectrum.00483-21 Google Scholar
Kittana, M, Ahmadani, A, Al Marzooq, F & Attlee, A (2021) Dietary fat effect on the gut microbiome, and its role in the modulation of gastrointestinal disorders in children with autism spectrum disorder. Nutrients 13(11), 3818.Google Scholar
Ma, B, Liang, J, Dai, M, Wang, J, Luo, J, Zhang, Z, et al. (2019) Altered gut microbiota in Chinese children with autism spectrum disorders. Front Cell Infect Microbiol [Internet]. Mar 6 [cited 2024 Jun 10]; 9. Available from: https://www.frontiersin.org/articles/10.3389/fcimb.2019.00040 Google Scholar
Xu, M, Xu, X, Li, J & Li, F (2019) Association between gut microbiota and autism spectrum disorder: a systematic review and meta-analysis. Front Psychiatry [Internet]. [cited 2024 Jun 10]; 10. Available from: https://www.frontiersin.org/journals/psychiatry/articles/10.3389/fpsyt.2019.00473/full Google Scholar
Bougafa, F (2023) Prevalence of H Pylori infection and related blood biomarker among autistic children in Tobruk City, Libya. AlQalam J Med Appl Sci 6(1), 274278.Google Scholar
Zhang, T, Zhu, X, Guo, J, Gu, AZ, Li, D & Chen, J (2021) Toxicity assessment of nano-ZnO exposure on the human intestinal microbiome, metabolic functions, and resistome using an in vitro colon simulator. Environ Sci Technol 55(10), 68846896.Google Scholar
Skrovanek, S, DiGuilio, K, Bailey, R, Huntington, W, Urbas, R, Mayilvaganan, B, et al. (2014) Zinc and gastrointestinal disease. World J Gastrointest Pathophysiol 5(4), 496513.Google Scholar
Xia, P, Lian, S, Wu, Y, Yan, L, Quan, G & Zhu, G (2021) Zinc is an important inter-kingdom signal between the host and microbe. Vet Res 52, 39.Google Scholar
Ulluwishewa, D, Anderson, RC, McNabb, WC, Moughan, PJ, Wells, JM & Roy, NC (2011) Regulation of tight junction permeability by intestinal bacteria and dietary components. J Nutr 141(5), 769776.Google Scholar
Zhong, W, McClain, CJ, Cave, M, Kang, YJ & Zhou, Z (2010) The role of zinc deficiency in alcohol-induced intestinal barrier dysfunction. Am J Physiol Gastrointest Liver Physiol 298(5), G625G633.Google Scholar
Al-Ayadhi, L, Zayed, N, Bhat, RS, Moubayed, NMS, Al-Muammar, MN & El-Ansary, A (2021) The use of biomarkers associated with leaky gut as a diagnostic tool for early intervention in autism spectrum disorder: a systematic review. Gut Pathogens 13(1), 54.Google Scholar
Kirsten, TB, Queiroz-Hazarbassanov, N, Bernardi, MM & Felicio, LF (2015) Prenatal zinc prevents communication impairments and BDNF disturbance in a rat model of autism induced by prenatal lipopolysaccharide exposure. Life Sci 130, 1217.Google Scholar
Kalyan, M, Tousif, AH, Sonali, S, Vichitra, C, Sunanda, T, Praveenraj, SS, et al. (2022) Role of endogenous lipopolysaccharides in neurological disorders. Cells 11(24), 4038.Google Scholar
Navarro, F, Pearson, DA, Fatheree, N, Mansour, R, Hashmi, SS & Rhoads, JM (2015) Are ‘leaky gut’ and behavior associated with gluten and dairy containing diet in children with autism spectrum disorders? Nutr Neurosci 18(4), 177185.Google Scholar
Yitik Tonkaz, G, Esin, IS, Turan, B, Uslu, H & Dursun, OB (2022) Determinants of leaky gut and gut microbiota differences in children with autism spectrum disorder and their siblings. J Autism Dev Disorders 53, 114.Google Scholar
de Magistris, L, Familiari, V, Pascotto, A, Sapone, A, Frolli, A, Iardino, P, et al. (2010) Alterations of the intestinal barrier in patients with autism spectrum disorders and in their first-degree relatives. J Pediatr Gastroenterol Nutr 51(4), 418424.Google Scholar
Fiorentino, M, Sapone, A, Senger, S, Camhi, SS, Kadzielski, SM, Buie, TM, et al. (2016) Blood–brain barrier and intestinal epithelial barrier alterations in autism spectrum disorders. Mol Autism 7(1), 49.Google Scholar
Sauer, AK & Grabrucker, AM (2019) Zinc deficiency during pregnancy leads to altered microbiome and elevated inflammatory markers in mice. Front Neurosci 13, 1295.Google Scholar
Gaulke, CA, Rolshoven, J, Wong, CP, Hudson, LG, Ho, E & Sharpton, TJ (2018) Marginal zinc deficiency and environmentally relevant concentrations of arsenic elicit combined effects on the gut microbiome. Msphere 3(6), e00521e00618.Google Scholar
Chen, X, Jiang, Y, Wang, Z, Chen, Y, Tang, S, Wang, S, et al. (2022) Alteration in gut microbiota associated with zinc deficiency in school-age children. Nutrients 14(14), 2895.Google Scholar
Sauer, AK, Hagmeyer, S & Grabrucker, AM (2022) Prenatal zinc deficient mice as a model for autism spectrum disorders. Int J Mol Sci 23(11), 6082.Google Scholar
Grabrucker, S, Boeckers, TM & Grabrucker, AM (2016) Gender dependent evaluation of autism like behavior in mice exposed to prenatal zinc deficiency. Front Behav Neurosci [Internet]. [cited 2022 Nov 16]; 10. Available from: https://www.frontiersin.org/articles/10.3389/fnbeh.2016.00037 Google Scholar
Grabrucker, S, Haderspeck, JC, Sauer, AK, Kittelberger, N, Asoglu, H, Abaei, A, et al. (2018) Brain lateralization in mice is associated with zinc signaling and altered in prenatal zinc deficient mice that display features of autism spectrum disorder. Front Mol Neurosci [Internet]. [cited 2022 Nov 16]; 10. Available from: https://www.frontiersin.org/articles/10.3389/fnmol.2017.00450 Google Scholar
Schoen, M, Asoglu, H, Bauer, HF, Müller, HP, Abaei, A, Sauer, AK, et al. (2019) Shank3 transgenic and prenatal zinc-deficient autism mouse models show convergent and individual alterations of brain structures in MRI. Front Neural Circuits [Internet]. [cited 2022 Nov 16]; 13. Available from: https://www.frontiersin.org/articles/10.3389/fncir.2019.00006 Google Scholar
Sauer, AK, Malijauskaite, S, Meleady, P, Boeckers, TM, McGourty, K & Grabrucker, AM (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.Google Scholar
Lin, P, Zhang, Q, Sun, J, Li, Q, Li, D, Zhu, M, et al. (2024) A comparison between children and adolescents with autism spectrum disorders and healthy controls in biomedical factors, trace elements, and microbiota biomarkers: a meta-analysis. Front Psychiatry [Internet]. Jan 9 [cited 2024 May 6]; 14. Available from: https://www.frontiersin.org/journals/psychiatry/articles/10.3389/fpsyt.2023.1318637/full Google Scholar
Benedictis, CAD, Haffke, C, Hagmeyer, S, Sauer, AK & Grabrucker, AM (2021) Expression analysis of zinc transporters in nervous tissue cells reveals neuronal and synaptic localization of ZIP4. Int J Mol Sci [Internet]. [cited 2022 Feb 18]; 22(9). Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8123391/ Google Scholar
Levaot, N & Hershfinkel, M (2018) How cellular Zn2+ signaling drives physiological functions. Cell Calcium 75, 5363.Google Scholar
Yoo, MH, Kim, TY, Yoon, YH & Koh, JY (2016) Autism phenotypes in ZnT3 null mice: involvement of zinc dyshomeostasis, MMP-9 activation and BDNF upregulation. Sci Rep 6(1), 115.Google Scholar
Pochwat, B, Nowak, G & Szewczyk, B (2015) Relationship between zinc (Zn2+) and glutamate receptors in the processes underlying neurodegeneration. Neural Plast 2015, 591563.Google Scholar
Frederickson, CJ, Suh, SW, Silva, D, Frederickson, CJ & Thompson, RB (2000) Importance of zinc in the central nervous system: the zinc-containing neuron. J Nutr 130(5), 1471S1483S.Google Scholar
Krall, RF, Moutal, A, Phillips, MB, Asraf, H, Johnson, JW, Khanna, R, et al. (2020) Synaptic zinc inhibition of NMDA receptors depends on the association of GluN2A with the zinc transporter ZnT1. Sci Adv [Internet]. Jul [cited 2022 Feb 5]. Available from: https://www.science.org/doi/abs/10.1126/sciadv.abb1515 Google Scholar
Sindreu, C, Palmiter, RD & Storm, DR (2011) Zinc transporter ZnT-3 regulates presynaptic Erk1/2 signaling and hippocampus-dependent memory. PNAS 108(8), 33663370.Google Scholar
Li, Y, Hough, CJ, Frederickson, CJ & Sarvey, JM (2001) Induction of mossy fiber→CA3 long-term potentiation requires translocation of synaptically released Zn2+. J Neurosci 21(20), 80158025.Google Scholar
Matias, CM, Dionísio, JC, Saggau, P & Quinta-Ferreira, ME (2014) Activation of group II metabotropic glutamate receptors blocks zinc release from hippocampal mossy fibers. Biol Res 47(1), 73.Google Scholar
Quinta-Ferreira, M, Sampaio dos Aidos, F, Matias, C, Mendes, P, Dionísio, J, Santos, R, et al. (2016) Modelling zinc changes at the hippocampal mossy fiber synaptic cleft. J Comput Neurosci 41, 323337.Google Scholar
Assaf, SY & Chung, SH (1984) Release of endogenous Zn2+ from brain tissue during activity. Nature 308(5961), 734736.Google Scholar
Rhodes, KJ, Strassle, BW, Monaghan, MM, Bekele-Arcuri, Z, Matos, MF & Trimmer, JS (1997) Association and colocalization of the Kvβ1 and Kvβ2 β-subunits with Kv1 α-subunits in mammalian brain K+ channel complexes. J Neurosci 17(21), 82468258.Google Scholar
Wang, H, Kunkel, DD, Schwartzkroin, PA & Tempel, BL (1994) Localization of Kv1. 1 and Kv1. 2, two K channel proteins, to synaptic terminals, somata, and dendrites in the mouse brain. J Neurosci 14(8), 45884599.Google Scholar
Cusimano, A, D’Adamo, MC & Pessia, M (2004) An episodic ataxia type-1 mutation in the S1 segment sensitises the hKv1. 1 potassium channel to extracellular Zn2+. FEBS Lett 576(1–2), 237244.Google Scholar
Imbrici, P, D’Adamo, MC, Cusimano, A & Pessia, M (2007) Episodic ataxia type 1 mutation F184C alters Zn2+-induced modulation of the human K+ channel Kv1. 4-Kv1. 1/Kvβ1. 1. Am J Physiol-Cell Physiol 292(2), C778C787.Google Scholar
Petrilli, MA, Kranz, TM, Kleinhaus, K, Joe, P, Getz, M, Johnson, P, et al. (2017) The emerging role for zinc in depression and psychosis. Front Pharmacol [Internet]. [cited 2022 Feb 5]; 8. Available from: https://www.frontiersin.org/article/10.3389/fphar.2017.00414 Google Scholar
Amico-Ruvio, SA, Murthy, SE, Smith, TP & Popescu, GK (2011) Zinc effects on NMDA receptor gating kinetics. Biophys J 100(8), 19101918.Google Scholar
Shi, R, Redman, P, Ghose, D, Hwang, H, Liu, Y, Ren, X, et al. (2017) Shank proteins differentially regulate synaptic transmission. eNeuro 4(6), ENEURO.0163-15.2017.Google Scholar
Uchino, S & Waga, C (2013) SHANK3 as an autism spectrum disorder-associated gene. Brain Dev 35(2), 106110.Google Scholar
Pfaender, S, Sauer, AK, Hagmeyer, S, Mangus, K, Linta, L, Liebau, S, et al. (2017) Zinc deficiency and low enterocyte zinc transporter expression in human patients with autism related mutations in SHANK3. Sci Rep 7(1), 115.Google Scholar
Monteiro, P & Feng, G (2017) SHANK proteins: roles at the synapse and in autism spectrum disorder. Nat Rev Neurosci 18(3), 147157.Google Scholar
Arons, MH, Lee, K, Thynne, CJ, Kim, SA, Schob, C, Kindler, S, et al. (2016) Shank3 is part of a zinc-sensitive signaling system that regulates excitatory synaptic strength. J Neurosci 36(35), 91249134.Google Scholar
Grabrucker, S, Jannetti, L, Eckert, M, Gaub, S, Chhabra, R, Pfaender, S, et al. (2014) Zinc deficiency dysregulates the synaptic ProSAP/Shank scaffold and might contribute to autism spectrum disorders. Brain 137(1), 137152.Google Scholar
Sauer, AK, Bockmann, J, Steinestel, K, Boeckers, TM & Grabrucker, AM (2019) Altered intestinal morphology and microbiota composition in the autism spectrum disorders associated SHANK3 mouse model. Int J Mol Sci 20(9), 2134.Google Scholar
Kim, Y, Ko, TH, Jin, C, Zhang, Y, Kang, HR, Ma, R, et al. (2023) The emerging roles of Shank3 in cardiac function and dysfunction. Front Cell Dev Biol 11, 1191369.Google Scholar
Lee, K, Jung, Y, Vyas, Y, Skelton, I, Abraham, WC, Hsueh, YP, et al. (2022) Dietary zinc supplementation rescues fear-based learning and synaptic function in the Tbr1+/− mouse model of autism spectrum disorders. Mol Autism 13(1), 13.Google Scholar
Bishop, SL & Lahvis, GP (2011) The autism diagnosis in translation: shared affect in children and mouse models of ASD. Autism Res 4(5), 317335.Google Scholar
Kazdoba, TM, Leach, PT & Crawley, JN (2016) Behavioral phenotypes of genetic mouse models of autism. Genes, Brain Behav 15(1), 726.Google Scholar
Tabouy, L, Getselter, D, Ziv, O, Karpuj, M, Tabouy, T, Lukic, I, et al. (2018) Dysbiosis of microbiome and probiotic treatment in a genetic model of autism spectrum disorders. Brain, Behav Immun 73, 310319.Google Scholar
Wei, SC, Yang-Yen, HF, Tsao, PN, Weng, MT, Tung, CC, Yu, LCH, et al. (2017) SHANK3 regulates intestinal barrier function through modulating ZO-1 expression through the PKCϵ-dependent pathway. Inflamm Bowel Dis 23(10), 17301740.Google Scholar
Wong, GC, Montgomery, JM & Taylor, MW (2021) The gut-microbiota-brain axis in autism spectrum disorder. In: Grabrucker, AM, editor. Autism Spectrum Disorders [Internet]. Brisbane, AU: Exon Publications; [cited 2022 Feb 17]. Available from: http://www.ncbi.nlm.nih.gov/books/NBK573606/ Google Scholar
Sgritta, M, Dooling, SW, Buffington, SA, Momin, EN, Francis, MB, Britton, RA, et al. (2019) Mechanisms underlying microbial-mediated changes in social behavior in mouse models of autism spectrum disorder. Neuron 101(2), 246259.e6.Google Scholar
Wong, GC, Jung, Y, Lee, K, Fourie, C, Handley, KM, Montgomery, JM, et al. (2021) Effect of dietary zinc supplementation on the gastrointestinal microbiota and host gene expression in the Shank3B−/− mouse model of autism spectrum disorder [Internet]. bioRxiv; [cited 2022 Feb 13]. Available from: https://www.biorxiv.org/content/10.1101/2021.09.09.459709v1 Google Scholar
Fourie, C, Vyas, Y, Lee, K, Jung, Y, Garner, CC & Montgomery, JM (2018) Dietary zinc supplementation prevents autism related behaviors and striatal synaptic dysfunction in Shank3 exon 13–16 mutant mice. Front Cell Neurosci 12, 374.Google Scholar
Mirzaei, R, Bouzari, B, Hosseini-Fard, SR, Mazaheri, M, Ahmadyousefi, Y, Abdi, M, et al. (2021) Role of microbiota-derived short-chain fatty acids in nervous system disorders. Biomed Pharmacother 139, 111661.Google Scholar
Grabrucker, AM (2014) A role for synaptic zinc in ProSAP/Shank PSD scaffold malformation in autism spectrum disorders. Dev Neurobiol 74(2), 136146.Google Scholar
Takeda, A (2000) Movement of zinc and its functional significance in the brain. Brain Res Rev 34(3), 137148.Google Scholar
Zhang, C, Dischler, A, Glover, K & Qin, Y (2022) Neuronal signalling of zinc: from detection and modulation to function. Open Biol 12(9), 220188.Google Scholar
Liu, X, Adamo, AM & Oteiza, PI (2023) Marginal zinc deficiency during gestation and lactation in rats affects oligodendrogenesis, motor performance, and behavior in the offspring. J Nutr 153(10), 27782796.Google Scholar
Wan, Y & Zhang, B (2022) The impact of zinc and zinc homeostasis on the intestinal mucosal barrier and intestinal diseases. Biomolecules 12(7), 900.Google Scholar
Halas, ES, Hunt, CD & Eberhardt, MJ (1986) Learning and memory disabilities in young adult rats from mildly zinc deficient dams. Physiol Behav 37(3), 451458.Google Scholar
Tahmasebi Boroujeni, S, Naghdi, N, Shahbazi, M, Farrokhi, A, Bagherzadeh, F, Kazemnejad, A, et al. (2009) The effect of severe zinc deficiency and zinc supplement on spatial learning and memory. Biol Trace Elem Res 130(1), 4861.Google Scholar
Sauer, AK, Hagmeyer, S & Grabrucker, AM (2016) Zinc deficiency. In Holick, M. F. & Nieves, J. W. (Eds.), Molecular, genetic, and nutritional aspects of major and trace minerals (pp. 2440) Rijeka, Croatia:IntechOpen.Google Scholar
Uriu-Adams, JY & Keen, CL (2010) Zinc and reproduction: effects of zinc deficiency on prenatal and early postnatal development. Birth Defects Res Part B: Dev Reprod Toxicol 89(4), 313325.Google Scholar
Indika, NLR, Frye, RE, Rossignol, DA, Owens, SC, Senarathne, UD, Grabrucker, AM, et al. (2023) The rationale for vitamin, mineral, and cofactor treatment in the precision medical care of autism spectrum disorder. J Personal Med 13(2), 252.Google Scholar
Irato, P, Sturniolo, GC, Giacon, G, Magro, A, D’Inca, R, Mestriner, C, et al. (1996) Effect of zinc supplementation on metallothionein, copper, and zinc concentration in various tissues of copper-loaded rats. Biol Trace Elem Res 51(1), 8796.Google Scholar
Vyas, Y, Lee, K, Jung, Y & Montgomery, JM (2020) Influence of maternal zinc supplementation on the development of autism-associated behavioural and synaptic deficits in offspring Shank3-knockout mice. Mol Brain 13(1), 110.Google Scholar
Vyas, Y, Jung, Y, Lee, K, Garner, CC & Montgomery, JM (2021) In vitro zinc supplementation alters synaptic deficits caused by autism spectrum disorder-associated Shank2 point mutations in hippocampal neurons. Mol Brain 14(1), 95.Google Scholar
Meyza, KZ, Defensor, EB, Jensen, AL, Corley, MJ, Pearson, BL, Pobbe, RL, et al. (2013) The BTBR T+ tf/J mouse model for autism spectrum disorders–in search of biomarkers. Behav Brain Res 251, 2534.Google Scholar
Zhang, L, Xu, X, Ma, L, Wang, X, Jin, M, Li, L, et al. (2023) Zinc water prevents autism-like behaviors in the BTBR mice. Biol Trace Elem Res 201(10), 47794792.Google Scholar
Figure 0

Figure 1. PRISMA flow diagram illustrating the search process, detailing the identification, screening, and quality assessment phases, which ultimately resulted in the inclusion of 224 studies and two reports in the final review.

Figure 1

Figure 2. Zn2+ metabolism in the human body. (a) Release of Zn2+ from nucleic and amino acid-bound complexes from food sources. (b) Zn2+ absorption through an enterocyte from the apical side through (1) ZIP4, (2) other routes, or (3) paracellular absorption. Zn2+ release from the enterocyte to the circulation occurs through ZnT1 transporter. In the blood, Zn2+ is carried by albumin (∼70%), macroglobulin (∼30%), or transferrin (∼10%). In the intracellular compartment, Zn2+ may be stored as part of metallothionein, which increases in Zn2+ supplementation and decreases with deficiency. (c) Zn2+ excretion through (1) feces, or (2) other routes.

Figure 2

Figure 3. Interplay between gut microbiota and Zn2+ to maintain a healthy gut. (a) In a Zn2+-abundant state, bacteria of the following phyla seem to thrive: Firmicutes, Actinobacteria, Verrucumicrobia, Proteobacteria, and Bacteroidetes. (b) In addition, the mucous membranes are intact with healthy enterocytes, and TJ. No inflammation or bacterial penetration is found. (c) In a Zn2+-deficient state, a decrease in Thermovirga and increase in Akkermansia, Blautia, Alloprevotella, and Ruminiclostridium were found in the cecum of mice, with an enrichment of Helicobacter hepaticus. (d) Enterocyte inflammation, impaired TJ, and proliferation of neutrophils along with pathogenic bacteria are found in a Zn2+-deficient state.

Figure 3

Figure 4. Crosstalk between microbiota-derived metabolites, their role in altering brain activity, and the potential impact of Zn2+ on the entire process (a, b). SCFAs may reach the CNS through the vagus nerve and play a neuroactive role (b). (c) Accumulation of toxic metabolites (lipopolysaccharides) produced by distinct gut bacteria that promote immune cell recruitment (d) and trigger the release of pro-inflammatory cytokines (e), could be key factors in the development of autistic phenotypes by affecting the synapses in the brain (f) via the gut–brain axis. (g) Zn2+ promotes neural communication and reduces inflammation by suppressing immune cell recruitment and reducing the production of pro-inflammatory cytokines. By contrast, low Zn2+ status can compromise intestinal barrier integrity and activate pro-inflammatory signaling, resulting in changes in microbiota composition that may aggravate inflammation.

Figure 4

Table 1 Effect of Zn2+ supplementation on ASD-related features or genes expression in individuals with ASD and animal and cell culture models of ASD