Hostname: page-component-848d4c4894-x5gtn Total loading time: 0 Render date: 2024-06-01T21:09:03.872Z Has data issue: false hasContentIssue false

Dysregulated phosphate metabolism in autism spectrum disorder: associations and insights for future research

Published online by Cambridge University Press:  13 June 2023

Ronald B. Brown*
Affiliation:
University of Waterloo, School of Public Health Sciences, Waterloo, ON N2L 3G1, Canada
*
Corresponding author: Ronald B. Brown Email: r26brown@uwaterloo.ca
Rights & Permissions [Opens in a new window]

Abstract

Studies of autism spectrum disorder (ASD) related to exposure to toxic levels of dietary phosphate are lacking. Phosphate toxicity from dysregulated phosphate metabolism can negatively impact almost every major organ system of the body, including the central nervous system. The present paper used a grounded theory-literature review method to synthesise associations of dysregulated phosphate metabolism with the aetiology of ASD. Cell signalling in autism has been linked to an altered balance between phosphoinositide kinases, which phosphorylate proteins, and the counteracting effect of phosphatases in neuronal membranes. Glial cell overgrowth in the developing ASD brain can lead to disturbances in neuro-circuitry, neuroinflammation and immune responses which are potentially related to excessive inorganic phosphate. The rise in ASD prevalence has been suggested to originate in changes to the gut microbiome from increasing consumption of additives in processed food, including phosphate additives. Ketogenic diets and dietary patterns that eliminate casein also reduce phosphate intake, which may account for many of the suggested benefits of these diets in children with ASD. Dysregulated phosphate metabolism is causatively linked to comorbid conditions associated with ASD such as cancer, tuberous sclerosis, mitochondrial dysfunction, diabetes, epilepsy, obesity, chronic kidney disease, tauopathy, cardiovascular disease and bone mineral disorders. Associations and proposals presented in this paper offer novel insights and directions for future research linking the aetiology of ASD with dysregulated phosphate metabolism and phosphate toxicity from excessive dietary phosphorus intake.

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

Introduction

The aetiology of autism spectrum disorder (ASD), a group of neurodevelopmental disorders that cause persistent social impairment with difficulties in communication and repetitive behavioural patterns, involves both genetic and environmental factors (Ref. Reference Hodges, Fealko and Soares1). Prevalence of ASD rapidly increased from 1 out of 150 U.S. children in 2000, to 1 out of 68 children in 2012 and 1 out of 59 children in 2018 (Ref. Reference Baio2). A recent systematic review and meta-analysis found that the male-to-female ratio of autism in children is approximately 3 to 1, which could be lower due to diagnostic gender bias that is less likely to diagnose females meeting ASD criteria compared to males (Ref. Reference Loomes, Hull and Mandy3). By 2016, black children in the United States were 1.5 times more likely to be identified with ASD than white or Hispanic children, suggesting that ASD is associated with social determinants of health involving socio-economic status, housing, physical environment and racial discrimination (Ref. Reference Shaw4).

Figure 1 shows clinical signs and symptoms of ASD, based on information provided by the American Academy of Pediatrics Council on Children With Disabilities (Ref. Reference Johnson and Myers5). Age of onset of ASD symptoms ranges from the first year of infancy up to 2–3 years (Ref. Reference Hyman, Levy and Myers6). Adolescents with ASD have a higher risk of developing depression, anxiety and attention-deficit hyperactivity disorder (Ref. Reference Accardo, Pontes and Pontes7). With no known cause, treatments for ASD patients are mostly symptomatic to improve quality of life and daily functioning (Ref. Reference Politte8).

Figure 1. Signs and symptoms of autism spectrum disorder, based on American Academy of Pediatrics Council on Children With Disabilities (Ref. Reference Johnson and Myers5).

Among potential environmental factors in ASD aetiology, dietary associations with ASD merit further investigations. In particular, studies of neurodevelopmental disorders from exposure to toxic levels of dietary phosphate are lacking. Dietary phosphorus in the form of inorganic phosphate (Pi) is an essential mineral that is regulated in the body by a sensitive network of hormones released by the kidneys, intestines, parathyroid glands and bone (Ref. Reference Brown and Razzaque9). Dysregulation of phosphate metabolism can lead to the accumulation of excess phosphate in the body causing a condition known as phosphate toxicity. Exposure to phosphate toxicity can negatively impact almost every major organ system in the body, including the central nervous system, with possible implications for the aetiology of neurodevelopmental disorders in ASD.

Method

The present narrative review used a grounded theory literature-review method (Ref. Reference Wolfswinkel, Furtmueller and Wilderom10) to search, retrieve and analyse research findings using keywords related to dysregulated phosphate metabolism, phosphate toxicity and ASD. Grounded theory in this method replaces subjectivity and conjecture with a rigorous method to synthesise new information grounded in evidence. Through an iterative process of comparative analysis, findings from the research literature were developed into themes and formed into associated relationships, and additional keywords were searched to follow the trail of evidence and fill in knowledge gaps. The strength of this method's bottom-up inductive approach lies in discovering potential breakthrough knowledge grounded in findings from published literature (Ref. Reference Brown11).

Nevertheless, ‘it is impossible to not be influenced by the background knowledge that one has’ (Ref. Reference Wolfswinkel, Furtmueller and Wilderom10), and limitations of this method, as in most other research methods, include potential selection and information biases that influence data selection, analysis, interpretation and presentation of the research findings (Ref. Reference Tripepi12). Findings may not be generalisable (external validity), and the researcher must successfully defend the work's internal validity under critical appraisal by peer experts. In the present paper, synthesised associations and proposals provide novel insights and directions for future research linking ASD aetiology and dysregulated phosphate metabolism.

ASD, cancer and phosphate

A retrospective cohort study of over 8000 children and adolescents with autistic disorder estimated that cancer incidence within the cohort was 94% higher than the expected cancer incidence (standardised incidence ratio 1.94) (Ref. Reference Chiang13). A more recent population-based study of 2.3 million individuals from Nordic countries found that overall risk from any cancer increased in ASD, but only in individuals with comorbid intellectual disability and/or birth defects (Ref. Reference Liu14), implying involvement of other causative factors such as chromosomal abnormalities related to mutations in PTEN (Phosphatase and Tensin Homolog), a tumour suppressor phosphatase (Ref. Reference Liu14). Phosphatases and protein kinases are enzymes that modulate cellular proteins by catalysing the transfer of phosphate in opposing directions (Ref. Reference Cheng15). A protein kinase catalyses the transfer of phosphate from ATP or GTP to phosphorylate a protein while a phosphatase catalyses the transfer of phosphate from a phosphoprotein to a water molecule. Similar to cell signalling in cancer (Ref. Reference Brown and Razzaque16), autism has been linked to an altered balance between phosphoinositide kinases and the counteracting effect of phosphatases in neuronal membranes (Ref. Reference Gross17).

Phosphorus is a growth-rate limiting factor in tumorigenesis (Ref. Reference Kuang, Nagy and Elser18). High dietary phosphate activates phosphoinositide 3-kinase (PI3 K) which phosphorylates Akt (protein kinase B) leading to activation of mTOR kinase that upregulates protein synthesis in cancer cell proliferation while suppressing apoptosis (Ref. Reference Guertin and Sabatini19). High dietary phosphate also suppresses the counteracting effect of PTEN on PI3 K (Ref. Reference Jin20). A similar cell-proliferation effect is seen in autism and epilepsy as PI3 K activates tuberous sclerosis complex 1/2 (TSC1/2) which activates mTOR kinase (Refs Reference Davis21, Reference Saxena and Sampson22). PTEN mutations in autism and epilepsy also suppress counteraction of PI3 K, leading to tumours and neurodevelopmental disorders in PTEN hamartoma tumour syndrome (Ref. Reference Yehia, Keel and Eng23). A study of brain analyses found that white matter in PTEN-ASD patients had a specific overgrowth pattern, with poor white matter development, reduced processing speed, increased deficits in working memory and extensive intellectual limitations (Ref. Reference Frazier24). Results of these brain tissue analyses imply that high concentrations of Pi may accumulate in the white matter of PTEN-ASD patients, who make up approximately 20% of children with ASD (Ref. Reference Busch25), and future studies should investigate Pi brain concentrations in PTEN-ASD patients.

Of relevance, phosphorus 31 magnetic resonance spectroscopy (MRS) was used in an earlier study to detect high concentrations of Pi in temporal lobe epilepsy (Ref. Reference Laxer26). More recently, increased Pi was detected in gliomas using MRS (Ref. Reference Peter and Nandhan27), and dysregulated expression of the gene PHD-finger protein 3 (PHF3) in glioblastoma overlaps with similar dysregulated gene expression of PHF3 in autism (Ref. Reference Appel28). Increased benign glioma risk of the central nervous system in children is also associated with neurofibromatosis type 1 (NF1) (Ref. Reference Costa and Gutmann29), and autistic behaviour is elevated in children with NF1 (Ref. Reference Chisholm30).

Feasibly, as in cancer, future research may show that high dietary phosphate associated with phosphate toxicity activates PI3 K and causes PTEN mutations in autism and epilepsy, leading to overgrowth and defective neuronal connectivity. Of relevance, phosphoric acid used as a common food additive was found to cause genotoxic damage to DNA in human lymphocytes (Ref. Reference Yilmaz31), which is an epigenetic factor that could increase PTEN mutations. Furthermore, lower urinary excretion of phosphoric acid was found in children with autism compared to controls (Ref. Reference Chen32), possibly indicating higher retention of phosphoric acid in body tissue with greater genotoxic damage in children with autism. More research is needed in the role of dysregulated phosphate metabolism in PTEN mutations in ASD.

ASD, dietary phosphate and gliosis

Infants who have been breastfed for long periods have better cognitive development and lower risk of autistic traits (Ref. Reference Boucher33). Phosphorus is about six-times lower in human breast milk (15 mg per 100 ml) than in cow milk (Ref. Reference Platt and Moncrieff34), and phosphorus plasma levels were lower and calcium levels higher in breastfed newborn babies compared to newborn babies fed unmodified cow milk (Ref. Reference Oppé and Redstone35). In infancy, high phosphorus, sodium, potassium, protein and chloride intake from inappropriate consumption of whole cow milk places a burden on developing kidneys' solute load (Ref. Reference Leung and Sauve36). These findings suggest that breastfeeding is associated with lower risk of renal burden and reduced incidence of dysregulated phosphate metabolism potentially related to autistic traits.

Casein in cow milk, a phosphoprotein with phosphate groups bonded to amino acid side groups, makes up about 80% of cow milk protein (Ref. Reference Sarode, Caballero, Finglas and Toldrá37). Accordingly, dietary patterns that reduce casein also reduce phosphorus, which may account for many of the suggested benefits of casein-free diets in children with ASD (Ref. Reference Quan38). Although gluten protein in grain does not contain phosphorus, bread and baked goods are often high in phosphorus and phosphate additives (Ref. Reference León, Sullivan and Sehgal39). While a negative effect from gluten ingestion remains unproven in ASD, gluten-free diets are suggested to have beneficial effects in ASD (Ref. Reference Croall, Hoggard and Hadjivassiliou40), which might be attributed to reductions in overall intake of phosphorus in bread and baked goods.

The rise in ASD has been suggested to originate in changes to the gut microbiome from increasing consumption of additives in processed food. Abdelli et al. found that exposing human neuron stem cells to the common food preservative propionic acid increased inflammatory responses and gliosis (excessive proliferation of fibrous support cells in the central nervous system) as occurs in neuro-circuitry dysfunction in ASD (Ref. Reference Abdelli, Samsam and Naser41). Interestingly, the researchers also found that propionic acid decreased levels of PTEN by about 50% compared to controls, which allowed levels of activated p-Akt (phosphorylated Akt) to increase and stimulate glial cell proliferation. Abdelli et al. also noted decreased neurite outgrowth in neuronal cells, which differentiate into dendrites or axons, and decreased axonal expansion was also noted which the researchers attributed to a physical barrier caused by excessive glia. The researchers proposed that dysregulated outgrowth and expansion explained disrupted communication between neuronal cells reported in ASD.

Figure 2 is a schematic diagram proposing that Pi from dysregulated phosphate metabolism phosphorylates Akt, leading to gliosis and neuro-circuitry disturbance in ASD. Future research should test this mechanism with in vitro experiments that compare glial cell proliferation in cultures exposed to concentrations of Pi, as well as in vivo experiments comparing levels of dietary phosphate and gliosis in lab animals. Although rodents are commonly used to model genetic mutations in autism, limited use of non-human primates comes closer to matching autistic behaviour in humans (Ref. Reference Katsnelson42).

Figure 2. Neuro-circuitry disturbance in autism spectrum disorder. Based on https://upload.wikimedia.org/wikipedia/commons/7/73/Blausen_0672_NeuralTissue.png.

ASD, dysregulated phosphate and immune/neuroinflammatory responses

Abdelli et al. measured immune responses and neuroinflammation in human neural stem cells from exposure to propionic acid, including increased levels of inflammatory cytokines such as tumour necrosis factor alpha, with smaller increases in the anti-inflammatory cytokine interleukin 10 (Ref. Reference Abdelli, Samsam and Naser41). Rodriguez and Kern proposed that reducing neuroinflammation and microglial activation could lead to improvements in neurodevelopment outcomes in people with ASD (Ref. Reference Rodriguez and Kern43).

Feasibly, immune responses related to neuro-circuitry disturbances in ASD may be caused by dysregulated phosphate. The following text compares abnormalities of natural killer cells (NK-cells), T-cells, B-cells, macrophages and monocytes in patients with ASD (Ref. Reference Gevezova44) with similar immune responses in conditions involving dysregulated phosphate. Although NK-cell proliferation was reported to increase in ASD, the NK-cells had reduced functioning ability (Ref. Reference Ashwood45). Normally, NK-cells regulate neuron and glia functions, including proliferation and neurite outgrowth (Ref. Reference Ebrahimi Meimand, Rostam-Abadi and Rezaei46), implying that NK-cell dysfunction and neuro-circuitry disturbances in ASD may share a common cause from exposure to phosphate toxicity. Of relevance, NK-cells are dysfunctional in end-stage kidney disease patients receiving dialysis therapy (Ref. Reference Nagai47), which is a procedure that helps control dysregulated phosphate (Ref. Reference Cupisti48).

T-cell abnormalities in ASD include reduced proliferation of T-helper cells (Th) and reduced regulatory T cells (Treg) with upregulation of Th2 cells (Ref. Reference Gevezova44). Interestingly, Th2 cells are involved in hypersensitivity to food antigens (Ref. Reference Ellenbogen49) which may include hypersensitivity from exposure to excessive dietary phosphate. Of relevance to increased skin sensitivity in ASD (Ref. Reference Jameson50), high serum phosphate is associated with skin itching—pruritus (Ref. Reference Swarna51), and pruritus is associated with increased Th2 cytokines (Ref. Reference Garcovich52). T cell exhaustion also occurs in kidney failure (Ref. Reference Hartzell53), which is associated with harmful levels of serum phosphate (Ref. Reference Rastogi54).

Children with ASD also have higher levels of pro-inflammatory B-cells [34]. B-cell infiltrates that occur in renal interstitial inflammation (Ref. Reference Heller55) are potentially related to B-cell infiltrates in kidney burden from phosphate toxicity (Ref. Reference Mironov, Atfi and Razzaque56). Macrophage and monocyte immune responses are also common in ASD (Ref. Reference Gevezova44) and in conditions with dysregulated phosphate such as kidney disease. For example, accumulation of macrophages and monocytes in renal tissue causes inflammation and is associated with renal tissue damage, scarring and reduced kidney function (Ref. Reference Wang57).

Dysregulated Pi in chronic kidney disease (CKD) also shares many other enzymes, immune responses and inflammatory markers with ASD. Compared to children with normal development, children with ASD were found to have elevated protein expression for IL-1β, IL-4, IL-9, interferon gamma (IFN-γ), Janus kinase 1 (JAK1), phosphorylated JAK1 (pJAK1), signal transducer and activator of transcription 5 (STAT5) and pSTAT5 (Ref. Reference Ahmad58). Similarly, patients with diabetic nephropathy, a leading cause of end-stage renal disease with dysregulated Pi, have increased expression of IL-1β and IL-4 (Ref. Reference Araújo59), and overproduction of IL-9 and IFN-γ was found in patients with acute glomerular injury of the kidneys (Ref. Reference Kim60). Additionally, JAK1 expression is enhanced in diabetic nephropathy (Ref. Reference Brosius and He61), phosphorylated JAK1 stimulates production of large amounts of FGF23, a regulator of phosphate metabolism (Ref. Reference Daryadel62), abnormal amounts of STAT5 are expressed in autosomal dominant polycystic kidney disease (Ref. Reference Fragiadaki63), and pSTAT5 is increased in focal segmental glomerulosclerosis of the kidneys (Ref. Reference Tao64).

Compared to children with normal development, children with ASD showed elevated expression of chemokine receptors in CD4 + T cells, including chemokine receptor 2 + (CXCR2 + ), CXCR3 + , CXCR5 +  and CXCR7 + and C-C motif chemokine receptor 3 + (CCR3 + ), CCR5 + , CCR7 +  and CCR9+ (Ref. Reference Ahmad65). Correspondingly, chemokine receptors CXCR1-7 are highly expressed in clear cell renal cell carcinoma (Ref. Reference Wu66), CCR3 is upregulated in renal cell carcinoma (Ref. Reference Jöhrer67), CCR5 + contributes to the pathogenesis of CKD (Ref. Reference Fuhro68), and children with CKD have increased expression of CCR7 and other chemokine receptors (Ref. Reference Szczepańska69). Furthermore, CCR9 + is associated with food allergens (Ref. Reference Castan70), which may include reactions to exposure to excessive dietary phosphate.

Nuclear factor-erythroid 2 related factor 2 (Nrf2), a transcription factor that protects immune cells against inflammation and oxidation, was reduced in children with autism (Ref. Reference Nadeem71). Similarly, impaired production of Nrf2 in CKD increases oxidation and inflammation leading to nephritis (Ref. Reference Ruiz72). Severity of symptoms in children with autism is associated with upregulation of IL-6 receptors and IL-17A in CD4 + T cells (Ref. Reference Nadeem73), and upregulated IL-17A expression in neutrophils increases inflammation and oxidation in children with autism (Ref. Reference Nadeem74). Likewise, IL-17A plays a central role in kidney disease pathogenesis (Ref. Reference Cortvrindt75) while IL-6 increases expression of FGF23 in acute and CKD (Ref. Reference Durlacher-Betzer76). Autism in children is also associated with elevated expression of IL-16 in CD4 + , CD8 + , CD14 + , CCR3 +  and CXCR7 + cells (Ref. Reference Ahmad77). Similarly, IL-16 contributes to inflammation of the kidneys which can lead to impaired kidney function (Ref. Reference Tavener, Jewell and Panickar78). Furthermore, immune dysfunction in children with autism is associated with upregulated T cell immunoglobulin and mucin domain 3 (TIM-3) (Ref. Reference Ahmad79), and overexpression of TIM-3 is associated with severe kidney inflammation (Ref. Reference Lu80).

Research is needed to explore common mechanisms that explain how dysregulated phosphate metabolism may contribute to immune and inflammatory responses in both kidney disease and ASD. Other environmental factors and toxic conditions can also contribute to ASD—nevertheless, evidence implicating dysregulated phosphate metabolism in ASD warrants further investigation.

ASD, phosphate additives and ultraprocessed food

U.S. consumption of ultra-processed foods, defined as industrially made, ready-to-eat or heat-and-serve products that contain food additives and generally lack whole foods, has continuously increased over the past two decades (Ref. Reference Juul81), concurrent with increases in ASD prevalence. Children with ASD often prefer highly processed foods with phosphate additives, including chicken nuggets, pizza, macaroni with cheese, breakfast cereals, pancakes, waffles, hot dogs, crackers and bread, with less preference for fresh produce (Ref. Reference Mayes and Zickgraf82). Compared to children with typical development, children with ASD were found to consume approximately 20–30% more ultra-processed food (Ref. Reference Buro, Kakkad and Gray83). Of relevance, consumption of ultraprocessed food high in inorganic phosphates is associated with renal function decline in older adults (Ref. Reference Rey-García84).

Additionally, a recent systematic review and meta-analysis found a 58% increased risk of obesity associated with ASD in children compared to controls (Ref. Reference Sammels85). Higher prevalence of childhood obesity in ASD may be related to higher energy intake and weight gain caused by ultra-processed food intake, as demonstrated in a controlled study of ultra-processed food consumed by adults with obesity (Ref. Reference Hall86). Maternal intake of ultra-processed food is also associated with increased obesity in offspring (Ref. Reference Wang87). Furthermore, a national cross-sectional study found that ultra-processed food consumption is greater among younger aged, lower income and less educated Americans (Ref. Reference Baraldi88), and more research is needed to compare these demographic findings with ASD prevalence. Because neurodevelopment in newborns is critically influenced by maternal prenatal diet, which is suggested to play a role in ASD aetiology (Ref. Reference Zhong89), research should investigate whether younger mothers with less education, lower income, obesity and greater consumption of ultra-processed food have a higher risk of ASD in their offspring.

Many sugar-sweetened beverages (SSBs), such as colas, contain phosphoric acid, and increased consumption of SSBs is associated with stronger and more difficult emotional problems in ASD (Ref. Reference Tan90). In addition, baking powder, chocolate, cocoa and beer are common sources of dietary phosphate that may contribute to excessive phosphate intake (Ref. Reference Picard91). A 2019 national survey of food items in Finland (representative of Europe) found phosphate additives in 36% of sampled foods, and 17 different phosphate additives were observed overall (Ref. Reference Tuominen, Karp and Itkonen92). Phosphorus-containing food additives are listed in Table 1.

Table 1. Phosphorus-containing food additives

Based on Tuominen et al. (Ref. Reference Tuominen, Karp and Itkonen92).

Sodium aluminium phosphate is commonly added to baking powder and processed cheese (Ref. Reference Saiyed and Yokel93). Excessive environmental exposure to aluminium within the body causes aluminium ions to bind with phosphate, which interferes with phosphorylation and dephosphorylation mechanisms in kinase and phosphatase enzymes, and contributes to mitochondrial dysfunction, microglia activation and immune and neuroinflammatory responses (Ref. Reference Morris, Puri and Frye94). Compared to controls, greater levels of aluminium in brain tissue have been detected in autism and neurodegenerative diseases (Ref. Reference Exley and Clarkson95), and intracellular aluminium in autism is concentrated within microglial cells and other non-neuron cells of brain tissue (Ref. Reference Mold96).

In addition to higher dietary phosphate intake, average concentrations of urinary phosphorus excretion are 34% lower in children with ASD (Ref. Reference Qureshi97), suggesting positive net phosphate absorption and tissue storage in ASD. Importantly, serum phosphate levels do not always accurately reflect tissue Pi storage levels (Ref. Reference Osuka and Razzaque98), and abnormal amounts of phosphate shifted from serum to tissue storage could explain hypophosphatemia found in some children with ASD compared to healthy children (Ref. Reference Afroz99). More studies are needed using phosphorus 31 MRS for detection of phosphate tissue storage in autism during brain development (Ref. Reference Minshew, Pettegrew and Garreau100).

Additionally, dietary fat is free of phosphorus, and lower phosphorus intake levels may explain benefits in ASD from high-fat ketogenic diets (Ref. Reference Li101). Ketogenic diets are effective in reducing or preventing seizures in epilepsy (Ref. Reference D'Andrea Meira102), and ASD is associated with epilepsy (Ref. Reference Besag103), implying that both ASD and epilepsy may share a common pathophysiological cause related to dysregulated phosphate metabolism and phosphate toxicity.

Evidence suggests that excessive dietary protein intake above optimal levels for neurodevelopment can be as harmful as deficient protein intake in perinatal care (Ref. Reference Barreault104). Of relevance, each gram of protein from dietary sources is directly related to approximately 12–14 mg phosphorus (Ref. Reference D'Alessandro, Piccoli and Cupisti105). Consequently, dietary protein intake levels above optimal amounts for neurodevelopment are likely to involve excessive amounts of phosphorus with increased risk of dysregulated phosphate metabolism.

Higher ASD prevalence in males is consistent with higher mean dietary phosphorus intake in U.S. male children aged 2–11 years compared to females, with significantly higher phosphorus intake in male adolescents aged 12–19 years (Ref. Reference Moshfegh, Kovalchik and Clemens106). Interestingly, some children with ASD show improvements in behaviour during fever (Ref. Reference Grzadzinski107). Coincidently, tumours also regress following fever (Ref. Reference Køstner108), and fever is associated with reduced appetite (Ref. 109), inferring lower food intake. Accordingly, reduction in dietary phosphate intake associated with fevers may act as a mediating factor that temporarily improves behaviours in ASD and may reduce tumours as lower phosphate levels limit the rate of cancer cell growth.

ASD, diabetes and dysregulated phosphate

Large studies have found that ASD is associated with increased prevalence of type 1 and type 2 diabetes mellitus (Ref. Reference Tromans110). Of relevance, neuronal degeneration and other complications in diabetes are associated with dysregulated phosphate metabolism (Ref. Reference Brown111). These findings infer that dysregulated phosphate metabolism is a pathophysiological mechanism common to diabetes mellitus and neuronal degeneration in ASD. Maternal diabetes in rats induces autism-like behaviour in offspring (Ref. Reference Wang112). In humans, maternal obesity comorbid with diabetes is associated with a greater risk of ASD in offspring compared to obesity or diabetes alone (Ref. Reference Li113). This finding implies that high dietary phosphate intake in obesity may interact with dysregulated phosphate metabolism in diabetes, leading to greater associated risk of ASD. Further research is needed to confirm an interactive relationship in ASD between obesity and diabetes mediated by excessive phosphate intake.

Prevalence of neurodevelopmental disorders, including ASD, is higher in children with type 1 diabetes, and diabetic nephropathy and retinopathy are also higher in type 1 diabetic children with neurodevelopment disorders (Ref. Reference Liu114). Of relevance, children with ASD have a higher risk of ophthalmologic diagnosis (Ref. Reference Chang115) and 25% of adults with ASD were found to have CKD (Ref. Reference Miot116), providing further support for dysregulated phosphate metabolism as a common pathophysiological cause in diabetic complications and ASD. Furthermore, low muscle tone or hypotonia is an established marker of individuals with ASD (Ref. Reference Gabis117). Similarly, hypotonia affects children with dysregulated phosphate metabolism in CKD (Ref. Reference Abd El Naby118), and hypotonia is associated with increased mortality in patients with renal failure receiving dialysis therapy for hyperphosphatemia (Ref. Reference Wanic-Kossowska and Czekalski119).

ASD, mitochondrial dysfunction and tauopathy

Mitochondrial dysfunction is associated with pathogenesis of ASD, and occurs in up to 80% of children with ASD (Ref. Reference Castora120). Coincidentally, mitochondrial dysfunction is associated with dysregulated phosphate metabolism in the pathogenesis of diabetes (Ref. Reference Brown111) and Parkinson's disease (Ref. Reference Brown121). Precipitates of calcium phosphate collect within the mitochondria matrix of pancreatic beta cells and substantia nigra cells that produce insulin and dopamine in diabetes and Parkinson's disease, respectively, which interfere with the function of complex I in the electron transfer chain during oxidative phosphorylation. The result of mitochondrial dysfunction in cells includes reduced ATP biosynthesis, apoptosis and cell death. Importantly, the majority of children with autism in an exploratory controlled study had below-normal values of complex 1 activity (Ref. Reference Giulivi122). Research is needed to examine the pathophysiologic effect of dysregulated phosphate metabolism, phosphate toxicity and calcium phosphate precipitation in mitochondrial dysfunction affecting neuronal cells of individuals with ASD.

In tauopathies associated with neurodegenerative diseases like Alzheimer disease and Parkinson's disease (Ref. Reference Zhang123), hyperphosphorylation of tau, a protein that assists in neuron cell maturation and cytoarchitecture, causes microtubule dysfunction which is linked with tau aggregation and neurofibrillary tangles (Ref. Reference Xia, Prokop and Giasson124). Autism-like behaviours have been linked to increased levels of frontotemporal lobe tau and neurofibrillary pathology in late-life dementia (Ref. Reference Rhodus125). Genetically lowering or removing tau in mouse models prevented or reduced key features of ASD, including abnormally enlarged head size, epilepsy and overactivation of the PI3 K/Akt/mTOR signalling pathway, which resulted in disinhibition of PTEN (Ref. Reference Tai126). Drugs that interfere with tau phosphorylation are proposed to treat tauopathies (Ref. Reference Xia, Prokop and Giasson124), but increased attention should focus on dietary interventions that reduce excessive phosphate intake, which may act as a rate-limiting factor to reduce tau hyperphosphorylation in the aetiology of neurodegenerative diseases and neurodevelopmental diseases like ASD. Importantly, hyperphosphorylation of tau is associated with glial-tau pathology in glial cells (Ref. Reference Kahlson and Colodner127), further implicating hyperphosphorylation as a cause of excessive glial cell proliferation and gliosis, which disrupts neuro-circuitry in ASD.

ASD, vitamin D, bone disorders and cardiovascular disease

The bioactive form of vitamin D, 1,25(OH)2D3, also known as calcitriol, is synthesised by the kidneys from the storage form of vitamin D, 25-hydroxy vitamin D, 25(OH)D3 (Ref. Reference Brown and Razzaque9). Calcitriol increases intestinal absorption of Pi during digestion, raising serum phosphorus levels as needed. To reduce excessive levels of serum Pi, the kidneys produce less bioactive vitamin D, which decreases phosphorus absorption in the intestines. Low levels of 25(OH)D3, are common in ASD (Ref. Reference Petruzzelli128), and lower serum levels of calcitriol and calcium were also found in ASD compared to normal controls (Ref. Reference Meguid129)—a potential biomarker indicating a kidney response in ASD that attempts to reduce serum Pi by lowering phosphate intestinal absorption when dietary phosphate intake is excessive.

Other endocrine hormones downregulate serum Pi by increasing renal phosphaturia, including fibroblast growth factor 23 (FGF23) released from bone and parathyroid hormone (PTH) released from the parathyroid glands (Ref. Reference Brown and Razzaque9). A recent case study reported elevated serum PTH and serum phosphate in a 14-year old boy with ASD having hypocalemic seizures following a prolonged diet of processed snack foods (Ref. Reference Kota, Kumar and Bargman130). Researchers also found structural changes in the frontal lobes associated with increased FGF23, likely in response to high levels of Pi which could also explain increased vascular calcification and microangiopathic lesions found in the brain leading to white matter loss (Ref. Reference Marebwa131). In general the brain's frontal lobes control social and cognitive behaviour, and frontal cortex damage is known to lead to the onset of persistent and repetitive behaviour, insistence for sameness, and impulsive behaviours which are clinical features of ASD (Ref. Reference Khadem-Reza and Zare132). Interestingly, low serum levels of basic fibroblast growth factor, fibroblast growth factor 2 (FGF2), which regulates neurodevelopment, were found in children with ASD (Ref. Reference Esnafoglu and Ayyıldız133). Future research is needed to investigate how dysregulated Pi impacts high FGF23 and low FGF2 levels in young children with ASD.

Odds of hip fractures in a case-control study were higher in children and adults with ASD compared to non-ASD individuals, and odds of fractures of the forearm and spine were greater in women with ASD (Ref. Reference Neumeyer134). Compared to typically developing children, a meta-analysis found that children with ASD had up to 13% lower bone mineral density of the total body, which is associated with increased risk of fractures (Ref. Reference Rostami Haji Abadi135). Osteoporosis and fracture are also associated with dysregulated phosphate metabolism in CKD-mineral and bone disease (CKD-MBD) (Ref. Reference Pazianas and Miller136), suggesting a common cause with fractures in ASD.

Bone mineral disorders related to dysregulated phosphate metabolism also impact poor oral health and periodontal disease, and dental plaque associated with gingivitis and periodontitis often forms from excessive calcium phosphate in saliva (Ref. Reference Brown137). A meta-analysis in children and young adults with ASD found a pooled prevalence of approximately 70% for periodontal disease (Ref. Reference da Silva138), implicating phosphate toxicity in ASD as a potential pathophysiological factor in poor oral health.

Autism is also associated with endothelial dysfunction of the arterial system, which contributes to disruption of neurovascular coupling mechanisms in neurodevelopment (Ref. Reference Ouellette139). In another potential link between ASD and dysregulated phosphate metabolism, hyperphosphatemia has a deleterious effect on endothelial dysfunction (Ref. Reference Stevens140). Of relevance, risk factors for cardiovascular disease (CVD) are increased in CKD, and the primary treatment goal in CKD is control of phosphate load (Ref. Reference Ritter and Slatopolsky141). Increased risk for heart failure and atrial fibrillation in CKD (Ref. Reference Rehm142) overlaps with similar risk for heart failure and atrial fibrillation in ASD (Ref. Reference Sun143). Moreover, a higher risk of death in people with coronary disease was associated with higher serum phosphate levels (Ref. Reference Tonelli144). Even people with normal renal function were found to have an increased risk of cardiovascular events associated with higher serum phosphate (Ref. Reference McGovern145). Although many environmental factors that contribute to ASD pathogenesis are shared with kidney disease, studies are lacking that investigate the association of paediatric kidney disease with ASD in children (Ref. Reference Clothier and Absoud146), especially related to dysregulated phosphate metabolism.

Directed acyclic graph

Figure 3 is a directed acyclic graph (DAG), often used in epidemiological and clinical research to illustrate causal pathways and mediating factors that help explain the association of outcomes with exposures to risk factors (Ref. Reference Digitale, Martin and Glymour147). The DAG in Figure 3 shows that dysregulated phosphate metabolism is a potential mediating factor causatively linking (solid arrows) associations between ASD and comorbidities reviewed in this paper (dotted arrow). The DAG helps identify new pathways mediated by dysregulated phosphate metabolism for future research in the cause and prevention of ASD and associated comorbidities.

Figure 3. Dysregulated phosphate metabolism is causatively linked to both autism spectrum disorder and comorbidities associated with autism spectrum disorder.

Conclusion

Evidence reviewed in this paper suggests that the effects of dysregulated phosphate metabolism and phosphate toxicity in ASD appear across the full lifespan, from the maternal stages of early development, throughout childhood and into adulthood. Dysregulated phosphate is proposed to stimulate gliosis of the brain in ASD, which disrupts neuro-circuitry by blocking axon extension and shortening neurite outgrowth. Children with ASD were found to have a wide variety of immune and neuroinflammatory responses. The rapid increase in ASD prevalence coincidences with the general population's increased consumption of ultraprocessed foods that are high in phosphate additives. Dysregulated phosphate metabolism is also causatively linked to comorbidities associated with ASD such as abnormal proliferation of cells, Akt kinase cell signalling and PTEN inhibition in cancer and tuberous sclerosis. Comorbidities also include calcium phosphate precipitation in mitochondrial dysfunction, impaired hormone secretion in diabetes, seizures in epilepsy, inflammatory and immune responses in CKD, obesity linked to excessive intake of ultraprocessed food, hyperphosphorylation in tauopathy, atrial fibrillation and heart failure in CVD, and bone resorption and ectopic calcification in bone mineral disorders.

Although other environmental factors and toxic conditions besides phosphate toxicity may contribute to ASD, findings implicating dysregulated phosphate metabolism in ASD warrant further investigation. The evidence presented in the present paper offers novel insights and directions for future research linking dysregulated phosphate metabolism with the aetiology of ASD. Additionally, future studies examining the effect of reduced dietary phosphorus in ASD are warranted.

Financial support

This research received no specific grant from any funding agency, commercial or not-for-profit sectors.

Conflict of interest

The author declares no conflict of interest.

References

Hodges, H, Fealko, C and Soares, N (2020) Autism spectrum disorder: definition, epidemiology, causes, and clinical evaluation. Translational Pediatrics 9(suppl. 1), S55S65.CrossRefGoogle ScholarPubMed
Baio, J et al. (2018) Prevalence of autism spectrum disorder among children aged 8 years - autism and developmental disabilities monitoring network, 11 sites, United States, 2014. MMWR. Surveillance Summaries 67, 123.CrossRefGoogle Scholar
Loomes, R, Hull, L and Mandy, WPL (2017) What is the male-to-female ratio in autism spectrum disorder? A systematic review and meta-analysis. Journal of the American Academy of Child & Adolescent Psychiatry 56, 466474.CrossRefGoogle ScholarPubMed
Shaw, KA et al. (2022) Progress and disparities in early identification of autism spectrum disorder: autism and developmental disabilities monitoring network, 2002–2016. Journal of the American Academy of Child & Adolescent Psychiatry 61, 905914.CrossRefGoogle ScholarPubMed
Johnson, CP and Myers, SM and American Academy of Pediatrics Council on Children With Disabilities (2007) Identification and evaluation of children with autism spectrum disorders. Pediatrics 120, 11831215.CrossRefGoogle ScholarPubMed
Hyman, SL, Levy, SE and Myers, SM (2020) Identification, evaluation, and management of children with autism spectrum disorder. Pediatrics 145, e20193447.Google ScholarPubMed
Accardo, AL, Pontes, NMH and Pontes, MCF (2022) Heightened anxiety and depression among autistic adolescents with ADHD: findings from the national survey of children's health 2016–2019. Journal of Autism and Developmental Disorders 3, 114.Google ScholarPubMed
Politte, LC et al. (2015) Evidence-based treatments for autism spectrum disorder. Current Treatment Options in Psychiatry 2, 3856.CrossRefGoogle Scholar
Brown, RB and Razzaque, MS (2015) Dysregulation of phosphate metabolism and conditions associated with phosphate toxicity. BoneKEy reports 4, 705.CrossRefGoogle ScholarPubMed
Wolfswinkel, JF, Furtmueller, E and Wilderom, CPM (2013) Using grounded theory as a method for rigorously reviewing literature. European Journal of Information Systems 22, 4555.CrossRefGoogle Scholar
Brown, RB (2020) Breakthrough knowledge synthesis in the age of Google. Philosophies 5, 4.CrossRefGoogle Scholar
Tripepi, G et al. (2010) Selection bias and information bias in clinical research. Nephron Clinical Practice 115, c94c99.CrossRefGoogle ScholarPubMed
Chiang, H-L et al. (2015) Risk of cancer in children, adolescents, and young adults with autistic disorder. The Journal of Pediatrics 166, 418423.e1.CrossRefGoogle Scholar
Liu, Q et al. (2022) Cancer risk in individuals with autism spectrum disorder. Annals of Oncology 33, 713719.CrossRefGoogle ScholarPubMed
Cheng, HC et al. (2011) Regulation and function of protein kinases and phosphatases. Enzyme Research 2011, 794089.CrossRefGoogle ScholarPubMed
Brown, RB and Razzaque, MS (2018) Phosphate toxicity and tumorigenesis. Biochimica et Biophysica Acta (BBA) – Reviews on Cancer 1869, 303309.CrossRefGoogle ScholarPubMed
Gross, C (2017) Defective phosphoinositide metabolism in autism. Journal of Neuroscience Research 95, 11611173.CrossRefGoogle ScholarPubMed
Kuang, Y, Nagy, JD and Elser, JJ (2004) Biological stoichiometry of tumor dynamics: mathematical models and analysis. Discrete and Continuous Dynamical Systems Series B 4, 221240.Google Scholar
Guertin, DA and Sabatini, DM (2007) Defining the role of mTOR in cancer. Cancer cell 12, 922.CrossRefGoogle ScholarPubMed
Jin, H et al. (2009) High dietary inorganic phosphate increases lung tumorigenesis and alters Akt signaling. American Journal of Respiratory and Critical Care Medicine 179, 5968.CrossRefGoogle ScholarPubMed
Davis, PE et al. (2015) Tuberous sclerosis: a new frontier in targeted treatment of autism. Neurotherapeutics: The Journal of the American Society for Experimental NeuroTherapeutics 12, 572583.CrossRefGoogle ScholarPubMed
Saxena, A and Sampson, JR (2015) Epilepsy in tuberous sclerosis: phenotypes, mechanisms, and treatments. Seminars in Neurology 35, 269276.Google ScholarPubMed
Yehia, L, Keel, E and Eng, C (2020) The clinical spectrum of PTEN mutations. Annual Review of Medicine 71, 103116.CrossRefGoogle ScholarPubMed
Frazier, TW et al. (2015) Molecular and phenotypic abnormalities in individuals with germline heterozygous PTEN mutations and autism. Molecular Psychiatry 20, 11321138.CrossRefGoogle ScholarPubMed
Busch, RM et al. (2019) Neurobehavioral phenotype of autism spectrum disorder associated with germline heterozygous mutations in PTEN. Translational Psychiatry 9, 253.CrossRefGoogle ScholarPubMed
Laxer, KD et al. (1992) Increased pH and seizure foci inorganic phosphate in temporal demonstrated by [31P]MRS. Epilepsia 33, 618623.CrossRefGoogle ScholarPubMed
Peter, SB and Nandhan, VR (2021) 31-Phosphorus magnetic resonance spectroscopy in evaluation of glioma and metastases in 3 T MRI. The Indian Journal of Radiology & Imaging 31, 873881.Google Scholar
Appel, LM et al. (2021) PHF3 regulates neuronal gene expression through the Pol II CTD reader domain SPOC. Nature Communications 12, 6078.CrossRefGoogle ScholarPubMed
Costa, AA and Gutmann, DH (2019) Brain tumors in neurofibromatosis type 1. Neuro-Oncology Advances 1, vdz040.Google ScholarPubMed
Chisholm, AK et al. (2022) Delineating the autistic phenotype in children with neurofibromatosis type 1. Molecular Autism 13, 3.CrossRefGoogle ScholarPubMed
Yilmaz, S et al. (2014) DNA damage in human lymphocytes exposed to four food additives in vitro. Toxicology and Industrial Health 30, 926937.CrossRefGoogle ScholarPubMed
Chen, Q et al. (2019) Urine organic acids as potential biomarkers for autism-Spectrum disorder in Chinese children. Frontiers in Cellular Neuroscience 13, 150.CrossRefGoogle ScholarPubMed
Boucher, O et al. (2017) Association between breastfeeding duration and cognitive development, autistic traits and ADHD symptoms: a multicenter study in Spain. Pediatric Research 81, 434442.CrossRefGoogle ScholarPubMed
Platt, BS and Moncrieff, A (1947) Nutritional comparison of human and cow's milk for infant-feeding. British Medical Bulletin 5, 177180.CrossRefGoogle Scholar
Oppé, T and Redstone, D (1968) Calcium and phosphorus levels in healthy newborn infants given various types of milk. The Lancet 291, 10451048.CrossRefGoogle Scholar
Leung, AK and Sauve, RS (2003) Whole cow's milk in infancy. Paediatrics & Child Health 8, 419421.CrossRefGoogle ScholarPubMed
Sarode, AR et al. (2016) Casein and caseinate: methods of manufacture. In Caballero, B, Finglas, PM and Toldrá, F (eds), Encyclopedia of Food and Health. Oxford: Academic Press, pp. 676682.CrossRefGoogle Scholar
Quan, L et al. (2021) A systematic review and meta-analysis of the benefits of a gluten-free diet and/or casein-free diet for children with autism spectrum disorder. Nutrition reviews 80, 12371246.CrossRefGoogle Scholar
León, JB, Sullivan, CM and Sehgal, AR (2013) The prevalence of phosphorus-containing food additives in top-selling foods in grocery stores. Journal of Renal Nutrition 23, 265270.e2.CrossRefGoogle ScholarPubMed
Croall, ID, Hoggard, N and Hadjivassiliou, M (2021) Gluten and autism spectrum disorder. Nutrients 13, 572.CrossRefGoogle ScholarPubMed
Abdelli, LS, Samsam, A and Naser, SA (2019) Propionic acid induces gliosis and neuro-inflammation through modulation of PTEN/AKT pathway in autism spectrum disorder. Scientific reports 9, 8824.CrossRefGoogle ScholarPubMed
Katsnelson, A (2018) Modeling autism. Lab Animal 47, 4144.CrossRefGoogle ScholarPubMed
Rodriguez, JI and Kern, JK (2011) Evidence of microglial activation in autism and its possible role in brain underconnectivity. Neuron Glia Biology 7, 205213.CrossRefGoogle ScholarPubMed
Gevezova, M et al. (2020) Inflammation and mitochondrial dysfunction in autism spectrum disorder. CNS & Neurological Disorders Drug Targets 19, 320333.CrossRefGoogle ScholarPubMed
Ashwood, P et al. (2011) In search of cellular immunophenotypes in the blood of children with autism. PloS one 6, e19299.CrossRefGoogle ScholarPubMed
Ebrahimi Meimand, S, Rostam-Abadi, Y and Rezaei, N (2021) Autism spectrum disorders and natural killer cells: a review on pathogenesis and treatment. Expert Review of Clinical Immunology 17, 2735.CrossRefGoogle ScholarPubMed
Nagai, K (2021) Dysfunction of natural killer cells in end-stage kidney disease on hemodialysis. Renal Replacement Therapy 7, 8.CrossRefGoogle ScholarPubMed
Cupisti, A et al. (2013) Phosphate control in dialysis. International Journal of Nephrology and Renovascular Disease 6, 193205.CrossRefGoogle ScholarPubMed
Ellenbogen, Y et al. (2018) The initiation of Th2 immunity towards food allergens. International Journal of Molecular Sciences 19, 1447.CrossRefGoogle ScholarPubMed
Jameson, C et al. (2022) Eczema and related atopic diseases are associated with increased symptom severity in children with autism spectrum disorder. Translational Psychiatry 12, 415.CrossRefGoogle ScholarPubMed
Swarna, SS et al. (2019) Pruritus associated with chronic kidney disease: a comprehensive literature review. Cureus 11, e5256.Google ScholarPubMed
Garcovich, S et al. (2021) Pruritus as a distinctive feature of type 2 inflammation. Vaccines (Basel) 9, 303.CrossRefGoogle ScholarPubMed
Hartzell, S et al. (2020) Kidney failure associates with T cell exhaustion and imbalanced follicular helper T cells. Frontiers in Immunology 11, 583702.CrossRefGoogle Scholar
Rastogi, A et al. (2021) Management of hyperphosphatemia in end-stage renal disease: a new paradigm. Journal of Renal Nutrition 31, 2134.CrossRefGoogle ScholarPubMed
Heller, F et al. (2007) The contribution of B cells to renal interstitial inflammation. American Journal of Pathology 170, 457468.CrossRefGoogle ScholarPubMed
Mironov, N, Atfi, A and Razzaque, MS (2022) Phosphate burden and organ dysfunction. Frontiers in Aging 3, 890985.CrossRefGoogle ScholarPubMed
Wang, X et al. (2021) The role of macrophages in kidney fibrosis. Frontiers in Physiology 12, 705838.Google ScholarPubMed
Ahmad, SF et al. (2017) Upregulation of IL-9 and JAK-STAT signaling pathway in children with autism. Progress in Neuro-Psychopharmacology & Biological Psychiatry 79, 472480.CrossRefGoogle ScholarPubMed
Araújo, LS et al. (2020) Renal expression of cytokines and chemokines in diabetic nephropathy. BMC Nephrology 21, 308.CrossRefGoogle ScholarPubMed
Kim, MG et al. (2022) Kidney VISTA prevents IFN-γ/IL-9 axis-mediated tubulointerstitial fibrosis after acute glomerular injury. Journal of Clinical Investigation 132, e151189.CrossRefGoogle ScholarPubMed
Brosius, FC III and He, JC (2015) JAK Inhibition and progressive kidney disease. Current Opinion in Nephrology and Hypertension 24, 8895.CrossRefGoogle ScholarPubMed
Daryadel, A et al. (2021) Systemic Jak1 activation provokes hepatic inflammation and imbalanced FGF23 production and cleavage. FASEB Journal 35, e21302.CrossRefGoogle ScholarPubMed
Fragiadaki, M et al. (2017) STAT5 drives abnormal proliferation in autosomal dominant polycystic kidney disease. Kidney International 91, 575586.CrossRefGoogle ScholarPubMed
Tao, J et al. (2018) JAK-STAT signaling is activated in the kidney and peripheral blood cells of patients with focal segmental glomerulosclerosis. Kidney International 94, 795808.CrossRefGoogle ScholarPubMed
Ahmad, SF et al. (2018) Upregulation of peripheral CXC and CC chemokine receptor expression on CD4(+) T cells is associated with immune dysregulation in children with autism. Progress in Neuro-Psychopharmacology & Biological Psychiatry 81, 211220.CrossRefGoogle ScholarPubMed
Wu, Z et al. (2020) Characterization of the prognostic values of the CXCR1-7 in clear cell renal cell carcinoma (ccRCC) microenvironment. Frontiers in Molecular Biosciences, 7, 601206.CrossRefGoogle ScholarPubMed
Jöhrer, K et al. (2005) Up-regulation of functional chemokine receptor CCR3 in human renal cell carcinoma. Clinical Cancer Research 11, 24592465.CrossRefGoogle ScholarPubMed
Fuhro, MI et al. (2022) The impact of intradialytic exercise on immune cells expressing CCR5 + in patients with chronic kidney disease: a cross-over trial. The International Journal of Artificial Organs 45, 221226.CrossRefGoogle ScholarPubMed
Szczepańska, M et al. (2015) Expression of chemokine receptors on peripheral blood T cells in children with chronic kidney disease. Mediators of Inflammation 2015, 536894.CrossRefGoogle Scholar
Castan, L et al. (2018) Food allergen-sensitized CCR9(+) lymphocytes enhance airways allergic inflammation in mice. Allergy 73, 15051514.CrossRefGoogle ScholarPubMed
Nadeem, A et al. (2020) Differential regulation of Nrf2 is linked to elevated inflammation and nitrative stress in monocytes of children with autism. Psychoneuroendocrinology 113, 104554.CrossRefGoogle ScholarPubMed
Ruiz, S et al. (2013) Targeting the transcription factor Nrf2 to ameliorate oxidative stress and inflammation in chronic kidney disease. Kidney International 83, 10291041.CrossRefGoogle ScholarPubMed
Nadeem, A et al. (2020) Dysregulation in IL-6 receptors is associated with upregulated IL-17A related signaling in CD4 + T cells of children with autism. Progress in Neuro-Psychopharmacology & Biological Psychiatry 97, 109783.CrossRefGoogle ScholarPubMed
Nadeem, A et al. (2019) Oxidative and inflammatory mediators are upregulated in neutrophils of autistic children: role of IL-17A receptor signaling. Progress in Neuro-Psychopharmacology & Biological Psychiatry 90, 204211.CrossRefGoogle ScholarPubMed
Cortvrindt, C et al. (2017) The role of interleukin-17A in the pathogenesis of kidney diseases. Pathology 49, 247258.CrossRefGoogle ScholarPubMed
Durlacher-Betzer, K et al. (2018) Interleukin-6 contributes to the increase in fibroblast growth factor 23 expression in acute and chronic kidney disease. Kidney International 94, 315325.CrossRefGoogle Scholar
Ahmad, SF et al. (2019) Elevated IL-16 expression is associated with development of immune dysfunction in children with autism. Psychopharmacology 236, 831838.CrossRefGoogle ScholarPubMed
Tavener, SK, Jewell, DE and Panickar, KS (2022) The increase in circulating levels of pro-inflammatory chemokines, cytokines, and complement C5 in canines with impaired kidney function. Current Issues in Molecular Biology 44, 16641676.CrossRefGoogle ScholarPubMed
Ahmad, SF et al. (2019) Dysregulation of T cell immunoglobulin and mucin domain 3 (TIM-3) signaling in peripheral immune cells is associated with immune dysfunction in autistic children. Molecular Immunology 106, 7786.CrossRefGoogle ScholarPubMed
Lu, C et al. (2021) An emerging role of TIM3 expression on T cells in chronic kidney inflammation. Frontiers in Immunology 12, 798683.CrossRefGoogle Scholar
Juul, F et al. (2021) Ultra-processed food consumption among US adults from 2001 to 2018. The American Journal of Clinical Nutrition 115, 211221.CrossRefGoogle Scholar
Mayes, SD and Zickgraf, H (2019) Atypical eating behaviors in children and adolescents with autism, ADHD, other disorders, and typical development. Research in Autism Spectrum Disorders 64, 7683.CrossRefGoogle Scholar
Buro, A, Kakkad, A and Gray, H (2020) P120 children with autism Spectrum disorder who are picky eaters may consume more ultra-processed foods than non-picky eaters. Journal of Nutrition Education and Behavior 52(suppl. 7), S73.CrossRefGoogle Scholar
Rey-García, J et al. (2021) Ultra-processed food consumption is associated with renal function decline in older adults: a prospective cohort study. Nutrients 13, 428.CrossRefGoogle ScholarPubMed
Sammels, O et al. (2022) Autism spectrum disorder and obesity in children: a systematic review and meta-analysis. Obesity Facts 15, 305320.CrossRefGoogle ScholarPubMed
Hall, KD et al. (2019) Ultra-processed diets cause excess calorie intake and weight gain: an inpatient randomized controlled trial of ad libitum food intake. Cell Metabolism 30, 6777.e3.CrossRefGoogle ScholarPubMed
Wang, Y et al. (2022) Maternal consumption of ultra-processed foods and subsequent risk of offspring overweight or obesity: results from three prospective cohort studies. BMJ 379, e071767.CrossRefGoogle ScholarPubMed
Baraldi, LG et al. (2018) Consumption of ultra-processed foods and associated sociodemographic factors in the USA between 2007 and 2012: evidence from a nationally representative cross-sectional study. BMJ Open 8, e020574.CrossRefGoogle ScholarPubMed
Zhong, C et al. (2020) Maternal dietary factors and the risk of autism spectrum disorders: a systematic review of existing evidence. Autism Research: Official Journal of the International Society for Autism Research 13, 16341658.CrossRefGoogle ScholarPubMed
Tan, S et al. (2022) The association between sugar-sweetened beverages and milk intake with emotional and behavioral problems in children with autism spectrum disorder. Frontiers in Nutrition 9, 927212.CrossRefGoogle ScholarPubMed
Picard, K et al. (2023) Currently available handouts for low phosphorus diets in chronic kidney disease continue to restrict plant proteins and minimally processed dairy products. Journal of Renal Nutrition 33, 4552.CrossRefGoogle ScholarPubMed
Tuominen, M, Karp, HJ and Itkonen, ST (2022) Phosphorus-Containing food additives in the food supply-an audit of products on supermarket shelves. Journal of Renal Nutrition 32, 3038.CrossRefGoogle ScholarPubMed
Saiyed, SM and Yokel, RA (2005) Aluminium content of some foods and food products in the USA, with aluminium food additives. Food Additives & Contaminants 22, 234244.CrossRefGoogle ScholarPubMed
Morris, G, Puri, BK and Frye, RE (2017) The putative role of environmental aluminium in the development of chronic neuropathology in adults and children. How strong is the evidence and what could be the mechanisms involved? Metabolic Brain Disease 32, 13351355.CrossRefGoogle ScholarPubMed
Exley, C and Clarkson, E (2020) Aluminium in human brain tissue from donors without neurodegenerative disease: a comparison with Alzheimer's disease, multiple sclerosis and autism. Scientific Reports 10, 7770.CrossRefGoogle ScholarPubMed
Mold, M et al. (2018) Aluminium in brain tissue in autism. Journal of Trace Elements in Medicine and Biology 46, 7682.CrossRefGoogle ScholarPubMed
Qureshi, F et al. (2020) Urinary essential elements of young children with autism spectrum disorder and their mothers. Research in Autism Spectrum Disorders 72, 101518.CrossRefGoogle ScholarPubMed
Osuka, S and Razzaque, MS (2012) Can features of phosphate toxicity appear in normophosphatemia? Journal of Bone and Mineral Metabolism 30, 1018.CrossRefGoogle ScholarPubMed
Afroz, S et al. (2020) Serum calcium and phosphate in children with autism spectrum disorder. Journal of Bangladesh Society of Physiologist 15, 7277.CrossRefGoogle Scholar
Minshew, NJ and Pettegrew, JW (1998) 31P Magnetic resonance spectroscopy and its application to autism and brain development. In Garreau, B (ed.), Neuroimaging in Child Neuropsychiatric Disorders. Berlin, Heidelberg: Springer Berlin Heidelberg, pp. 181197.CrossRefGoogle Scholar
Li, Q et al. (2021) A ketogenic diet and the treatment of autism spectrum disorder. Frontiers in Pediatrics 9, 650624.Google ScholarPubMed
D'Andrea Meira, I et al. (2019) Ketogenic diet and epilepsy: what we know so far. Frontiers in Neuroscience 13, 5.CrossRefGoogle ScholarPubMed
Besag, FM (2018) Epilepsy in patients with autism: links, risks and treatment challenges. Neuropsychiatric Disease and Treatment 14, 110.CrossRefGoogle ScholarPubMed
Barreault, S et al. (2019) Impact of early protein and energy intakes on neurodevelopment at 2 years of corrected age in very low birth weight infants: a single-center observational study. PLoS ONE 14, e0218887.CrossRefGoogle ScholarPubMed
D'Alessandro, C, Piccoli, GB and Cupisti, A (2015) The “phosphorus pyramid”: a visual tool for dietary phosphate management in dialysis and CKD patients. BMC Nephrology 16, 9.CrossRefGoogle ScholarPubMed
Moshfegh, A, Kovalchik, A and Clemens, J (2016) Phosphorus intake of Americans: what we eat in America, NHANES 2011–2012. Food Surveys Research Group Dietary Data Brief No 15.Google Scholar
Grzadzinski, R et al. (2018) Children with autism spectrum disorder who improve with fever: insights from the simons simplex collection. Autism Research: Official Journal of the International Society for Autism Research 11, 175184.CrossRefGoogle ScholarPubMed
Køstner, AH et al. (2013) Regression in cancer following fever and acute infection. Acta Oncologica 52, 455457.CrossRefGoogle ScholarPubMed
IOM (1993) Nutritional needs in hot environments: Applications for military personnel in field operations. Institute of Medicine (US) Committee on Military Nutrition Research. 1993. In.Google Scholar
Tromans, S et al. (2020) The prevalence of diabetes in autistic persons: a systematic review. Clinical Practice and Epidemiology in Mental Health 16, 212225.CrossRefGoogle ScholarPubMed
Brown, RB (2020) Diabetes, diabetic complications, and phosphate toxicity: a scoping review. Current Diabetes Reviews 16, 674–689.CrossRefGoogle ScholarPubMed
Wang, X et al. (2019) Maternal diabetes induces autism-like behavior by hyperglycemia-mediated persistent oxidative stress and suppression of superoxide dismutase 2. Proceedings of the National Academy of Sciences of the USA 116, 2374323752.CrossRefGoogle ScholarPubMed
Li, M et al. (2016) The association of maternal obesity and diabetes with autism and other developmental disabilities. Pediatrics 137, e20152206.CrossRefGoogle ScholarPubMed
Liu, S et al. (2021) Neurodevelopmental disorders, glycemic control, and diabetic complications in type 1 diabetes: a nationwide cohort study. The Journal of Clinical Endocrinology & Metabolism 106, e4459e4e70.CrossRefGoogle ScholarPubMed
Chang, MY et al. (2021) Prevalence of ophthalmologic diagnoses in children with autism spectrum disorder using the optum dataset: aPopulation-based study. American Journal of Ophthalmology 221, 147153.CrossRefGoogle ScholarPubMed
Miot, S et al. (2019) Comorbidity burden in adults with autism spectrum disorders and intellectual disabilities – A report from the EFAAR (frailty assessment in ageing adults with autism spectrum and intellectual disabilities) study. Frontiers in Psychiatry 10, 00617.CrossRefGoogle ScholarPubMed
Gabis, LV et al. (2021) The weak link: hypotonia in infancy and autism early identification. Frontiers in Neurology 12, 612674.CrossRefGoogle ScholarPubMed
Abd El Naby, SA et al. (2020) Neurophysiological and neuroradiological changes in children with chronic kidney disease. Frontiers in Pediatrics 8, 570708.CrossRefGoogle ScholarPubMed
Wanic-Kossowska, M and Czekalski, S (2007) [Hypotonia in renal failure patients undergoing dialysis therapy]. Polskie Archiwum Medycyny Wewnetrznej 117, 5863.Google ScholarPubMed
Castora, FJ (2019) Mitochondrial function and abnormalities implicated in the pathogenesis of ASD. Progress in Neuro-Psychopharmacology and Biological Psychiatry 92, 83108.CrossRefGoogle ScholarPubMed
Brown, RB (2022) Parkinson's disease etiology: insights and associations with phosphate toxicity. International Journal of Molecular Sciences 23, 8060.CrossRefGoogle ScholarPubMed
Giulivi, C et al. (2010) Mitochondrial dysfunction in autism. JAMA 304, 23892396.CrossRefGoogle ScholarPubMed
Zhang, X et al. (2018) Tau pathology in Parkinson's disease. Frontiers in Neurology 9, 809.CrossRefGoogle ScholarPubMed
Xia, Y, Prokop, S and Giasson, BI (2021) “Don't Phos over Tau”: recent developments in clinical biomarkers and therapies targeting tau phosphorylation in Alzheimer's disease and other tauopathies. Molecular Neurodegeneration 16, 37.CrossRefGoogle ScholarPubMed
Rhodus, EK et al. (2022) Frontotemporal neurofibrillary tangles and cerebrovascular lesions are associated with autism spectrum behaviors in late-life dementia. Journal of Neurology 269, 51055113.CrossRefGoogle ScholarPubMed
Tai, C et al. (2020) Tau reduction prevents key features of autism in mouse models. Neuron 106, 421437.e11.CrossRefGoogle ScholarPubMed
Kahlson, MA and Colodner, KJ (2015) Glial Tau pathology in tauopathies: functional consequences. Journal of Experimental Neuroscience 9, 4350.Google ScholarPubMed
Petruzzelli, MG et al. (2020) Vitamin D deficiency in autism spectrum disorder: a cross-sectional study. Disease Markers 2020, 9292560.CrossRefGoogle ScholarPubMed
Meguid, NA et al. (2010) Reduced serum levels of 25-hydroxy and 1,25-dihydroxy vitamin D in Egyptian children with autism. Journal of Alternative and Complementary Medicine 16, 641645.CrossRefGoogle ScholarPubMed
Kota, A, Kumar, K and Bargman, R (2019) Hypocalcemic seizures in an adolescent with autism spectrum disorder. Consultant360.com. 2019. In.Google Scholar
Marebwa, BK et al. (2018) Fibroblast growth factor23 is associated with axonal integrity and neural network architecture in the human frontal lobes. PLoS ONE 13, e0203460.CrossRefGoogle ScholarPubMed
Khadem-Reza, ZK and Zare, H (2022) Evaluation of brain structure abnormalities in children with autism spectrum disorder (ASD) using structural magnetic resonance imaging. The Egyptian Journal of Neurology, Psychiatry and Neurosurgery 58, 135.CrossRefGoogle Scholar
Esnafoglu, E and Ayyıldız, SN (2017) Decreased levels of serum fibroblast growth factor-2 in children with autism spectrum disorder. Psychiatry Research 257, 7983.CrossRefGoogle ScholarPubMed
Neumeyer, AM et al. (2015) Brief report: bone fractures in children and adults with autism spectrum disorders. Journal of Autism and Developmental Disorders 45, 881887.CrossRefGoogle ScholarPubMed
Rostami Haji Abadi, M et al. (2021) Bone health in children and youth with ASD: a systematic review and meta-analysis. Osteoporosis International: A Journal Established as Result of Cooperation between the European Foundation for Osteoporosis and the National Osteoporosis Foundation of the USA 32, 16791691.CrossRefGoogle ScholarPubMed
Pazianas, M and Miller, PD (2021) Osteoporosis and chronic kidney disease-mineral and bone disorder (CKD-MBD): back to basics. American Journal of Kidney Diseases: The Official Journal of the National Kidney Foundation 78, 582589.CrossRefGoogle ScholarPubMed
Brown, RB (2019) Dysregulated phosphate metabolism, periodontal disease, and cancer: possible global health implications. Dentistry Journal 7, 18.CrossRefGoogle ScholarPubMed
da Silva, SN et al. (2017) Oral health status of children and young adults with autism spectrum disorders: systematic review and meta-analysis. International Journal of Paediatric Dentistry 27, 388398.CrossRefGoogle Scholar
Ouellette, J et al. (2020) Vascular contributions to 16p11.2 deletion autism syndrome modeled in mice. Nature Neuroscience 23, 10901101.CrossRefGoogle ScholarPubMed
Stevens, KK et al. (2017) Deleterious effects of phosphate on vascular and endothelial function via disruption to the nitric oxide pathway. Nephrology Dialysis Transplantation 32, 16171627.Google Scholar
Ritter, CS and Slatopolsky, E (2016) Phosphate toxicity in CKD: the killer among us. Clinical Journal of the American Society of Nephrology 11, 10881100.CrossRefGoogle ScholarPubMed
Rehm, M et al. (2022) Chronic kidney disease and risk of atrial fibrillation and heart failure in general population-based cohorts: the BiomarCaRE project. ESC Heart Failure 9, 5765.CrossRefGoogle ScholarPubMed
Sun, X et al. (2021) Association of autism spectrum disorder, neuroticism, and subjective well-being with cardiovascular diseases: a two-sample Mendelian randomization study. Frontiers in Cardiovascular Medicine 8, 676030.CrossRefGoogle ScholarPubMed
Tonelli, M et al. (2005) Relation between serum phosphate level and cardiovascular event rate in people with coronary disease. Circulation 112, 26272633.CrossRefGoogle ScholarPubMed
McGovern, AP et al. (2013) Serum phosphate as a risk factor for cardiovascular events in people with and without chronic kidney disease: a large community based cohort study. PLoS ONE 8, e74996.CrossRefGoogle ScholarPubMed
Clothier, J and Absoud, M (2021) Autism spectrum disorder and kidney disease. Pediatric Nephrology 36, 29872995.CrossRefGoogle ScholarPubMed
Digitale, JC, Martin, JN and Glymour, MM (2022) Tutorial on directed acyclic graphs. Journal of Clinical Epidemiology 142, 264267.CrossRefGoogle ScholarPubMed
Figure 0

Figure 1. Signs and symptoms of autism spectrum disorder, based on American Academy of Pediatrics Council on Children With Disabilities (Ref. 5).

Figure 1

Figure 2. Neuro-circuitry disturbance in autism spectrum disorder. Based on https://upload.wikimedia.org/wikipedia/commons/7/73/Blausen_0672_NeuralTissue.png.

Figure 2

Table 1. Phosphorus-containing food additives

Figure 3

Figure 3. Dysregulated phosphate metabolism is causatively linked to both autism spectrum disorder and comorbidities associated with autism spectrum disorder.