Next Article in Journal
Normative Data for the D-KEFS Color-Word Interference and Trail Making Tests Adapted in Greek Adult Population 20–49 Years Old
Previous Article in Journal
Sensory Processing Challenges in Children with Neurodevelopmental Disorders and Genetic Conditions: An Observational Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Viewpoint

The Intersection of Ultra-Processed Foods, Neuropsychiatric Disorders, and Neurolaw: Implications for Criminal Justice

by
Susan L. Prescott
1,2,3,
Kathleen F. Holton
4,
Christopher A. Lowry
5,
Jeffrey J. Nicholson
6 and
Alan C. Logan
1,*
1
Nova Institute for Health, Baltimore, MD 21231, USA
2
School of Medicine, University of Western Australia, Perth, WA 6009, Australia
3
Department of Family and Community Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
4
Departments of Health Studies and Neuroscience, American University, Washington, DC 20016, USA
5
Department of Integrative Physiology, Department of Psychology and Neuroscience, Center for Neuroscience, and Center for Microbial Exploration, University of Colorado Boulder, Boulder, CO 80309, USA
6
Law and Government, Humber College Institute of Technology & Advanced Learning, Toronto, ON M9W 5L7, Canada
*
Author to whom correspondence should be addressed.
NeuroSci 2024, 5(3), 354-377; https://doi.org/10.3390/neurosci5030028
Submission received: 17 August 2024 / Revised: 18 September 2024 / Accepted: 19 September 2024 / Published: 23 September 2024

Abstract

:
Over the last decade there has been increasing interest in the links between the consumption of ultra-processed foods and various neuropsychiatric disorders, aggression, and antisocial behavior. Neurolaw is an interdisciplinary field that seeks to translate the rapid and voluminous advances in brain science into legal decisions and policy. An enhanced understanding of biophysiological mechanisms by which ultra-processed foods influence brain and behavior allows for a historical reexamination of one of forensic neuropsychiatry’s most famous cases—The People v. White and its associated ‘Twinkie Defense’. Here in this Viewpoint article, we pair original court transcripts with emergent research in neurolaw, including nutritional neuroscience, microbiome sciences (legalome), pre-clinical mechanistic research, and clinical intervention trials. Advances in neuroscience, and related fields such as the microbiome, are challenging basic assumptions in the criminal justice system, including notions of universal free will. Recent dismissals of criminal charges related to auto-brewery syndrome demonstrate that courts are open to advances at the intersection of neuromicrobiology and nutritional neuroscience, including those that relate to criminal intent and diminished capacity. As such, it is our contention that experts in the neurosciences will play an increasing role in shaping research that underpins 21st-century courtroom discourse, policy, and decision-making.

1. Introduction

The notoriety of the Twinkie defense may not soon be rivalled…but in a system of justice based on responsibility and intentionality, the [neuropsychiatric] discoveries raise troubling questions about the morality of punishing those who may lack awareness that they are sick.
Kirk Makin, Legal Affairs Journalist, 1988 [1]
Neuroscientists and neuropsychologists are often called upon to provide expert testimony, especially as courts warm to scientific advances within the brain sciences [2,3]. Experts in the field assist the courts in making determinations of competency and capacity [4], including a defendant’s decision-making capacity in relation to criminal culpability [5]. In the United States, the rate of traumatic brain injury among prisoners is five times higher than the general population [6], while a recent European study using brain imaging showed that half of inmates displayed signs of brain pathology compared to 8% among healthy non-criminal controls [7]. The odds of criminal justice involvement among veterans with posttraumatic stress disorder (PTSD) are 61% higher than those without PTSD, and the odds of arrest for violent offenses are 59% higher [8]. Evidence shows that neuropsychiatric conditions are often criminalized [9]. The interdisciplinary sector that attempts to translate the rapid and voluminous advances in brain science into legal decisions and policy is known as neurolaw [10]. Neurolaw encompasses neurology, neuropsychiatry, neuroimaging, neurogenetics, neuro-microbiology, forensic neuropsychology, and related disciplines [11,12].
In the context of a criminal justice system that is based on universal assumptions related to free will and criminal intent, advances in neuroscience have profound implications for individuals, communities, and society writ-large [13,14]. For example, developmental neuroscience has upended assumptions related to sharp delineations between the adolescent and adult brain, and these findings have led to changes in the ways in which teenagers are treated by the courts [15]. A growing awareness of emerging adulthood as a distinct phase of development may have future implications. A part of ongoing discussions within neurolaw is the extent to which emerging findings require a greater responsiveness by the courts, including acceptance that criminal responsibility and blameworthiness do not sit on an even plane [16]. In addition, findings in the realm of forensic neurosciences can aid in understanding offender risk and best practices for rehabilitation [17]. However, advances in neuroscience and other biomedical sciences are often siloed away from the criminal justice field, and the relevancy of findings to the justice system is often underappreciated or relegated in importance [18,19].
Although the term neurolaw emerged in the early 1990s, tethered to personal injury lawsuits and the application of validated neurocognitive testing [20,21], the use of objective tests (e.g., electroencephalogram (EEG)) to limit criminal liability dates back to the 1930s [11,22]. Although relatively rare, defense teams have won acquittals (or lesser or abandonment of charges) when using neuroimaging and related assessments as part of the defense strategy [23]. Among the exciting advances in neurosciences as they relate to antisocial, aggressive, and violent behavior, is the realm of nutritional neuroscience [24,25,26,27] and its intersection with the microbiome [28,29]. In the context of human cognition and behavior, emerging research demonstrates a bidirectional relationship between dietary patterns and the microbiome [27]. That is, dietary choices influence microbiota, which, via the bidirectional microbiome–gut–brain axis, influence the brain. At the same time, microbiota appear to influence nutritional status, and perhaps even dietary-related behaviors. Neuroscience, as informed by emergent microbiome findings, provides an important mechanistic understanding of epidemiological links between diet and behavior.
Here in this viewpoint article, we provide a retrospective analysis of one of the most famous cases in forensic neuropsychiatry, The People v. White, and its resultant ‘Twinkie Defense’. We use original court testimony and draw from emergent research in the fields of nutritional neuroscience [26] and the microbiome in forensics (i.e., the legalome) [18]. It is our contention that the synthesis of cutting-edge research from the top-down (nutritional epidemiology), bottom-up (mechanistic pre-clinical studies and microbiome sciences), and head-on (human intervention studies) perspectives, provides an entirely new outlook on the role of nutrition and the microbiome in forensic neurosciences. Moreover, the emergent research on ultra-processed food addiction has raised important legal questions (including individual diminished capacity and corporate culpability) under the umbrella term of food crime [30,31]. Although discussions of the Twinkie Defense are often ahistorical, we consider the broad contextual history to be an important part of its relationship to contemporary findings. That is, the Twinkie Defense did not emerge from a vacuum. Rather, it emerged from the forensic neuroscience ‘battlegrounds’ that marked the beginning of the end of Freudian pseudoscience [32,33] and untestable, subjective psychoanalytic dogma that plagued the courts [34].
Drawing from the PsycINFO, Google Scholar, and PubMed databases, we revisit the defense of diminished capacity through the lens of contemporary scientific research. We supplement the material found in academic databases with relevant articles drawn from media databases, including NewsBank and Ancestry’s Newspapers.com. This history and emergent evidence are pertinent because, as discussed below, a variation of the Twinkie Defense has recently proved successful in driving while intoxicated (DWI) cases in the United States and Europe [18].

2. The Twinkie Defense

On 27 November 1978, Daniel J. White, a disgruntled former city of San Francisco employee, used a handgun to fatally injure the mayor of San Francisco, George Moscone, and city supervisor, Harvey Milk. Shortly after the incident, White surrendered to local law enforcement. Despite his surrender and confession, White’s case went to trial with a defense of diminished capacity—an impairment not amounting to insanity, but one capable of removing the “malice aforethought” and other pre-commission mental states essential to murder. In California law, the focus was on “reduced mental capacity, whether caused by mental illness, mental defect, intoxication, or any other” [35]. White was charged with murder and a potential death penalty awaited him at sentencing. However, in White’s case, the diminished capacity defense was successful, as the jury found him guilty of voluntary manslaughter.
The basis of White’s defense was that he was suffering from depression wherein an unhealthy diet was not merely an association, it was a contributor to his altered mental state. At trial, White’s unhealthy dietary pattern was discussed multiple times, and as we will discuss below, it was presented as a factor of causation in his cognition and behavior. For the media reporting on the trial in real time, White’s claimed dietary pattern was encapsulated in the mention, by both the prosecution and the defense, of a specific highly processed food-like product—Twinkies. Perhaps because of the product’s broad significance in American culture, Twinkies became representative of the larger aspects of an ultra-processed dietary pattern, and the defense strategy was dubbed ‘The Twinkie Defense’.
Many contemporary articles repeat the notion that the defense team barely mentioned junk food as part of the defense strategy, and that insofar as junk food was presented as a causative factor in diminished capacity, the Twinkie Defense is claimed to be a “myth” [36,37]. Even textbooks in the category of forensics claim that White’s diminished capacity defense was exclusively about depression and the verdict had nothing to do with Twinkies or any such ultra-processed foods [38,39]. In a multi-author academic text on testimony from brain science experts, it is claimed that “The trial for double-murder concluded without reference to White’s poor eating habits. It was not until after the jury’s verdict and the judge’s sentencing were made public that the issue of sugar consumption as an influence on White’s criminal behavior was raised” [40]. One of the most highly regarded books in biological criminology repeats this same false claim that Twinkies “were never actually brought up at Dan White’s trial” [41]. Retrospective articles in high profile news outlets also claim that a mere “throwaway remark” about junk food by the defense, led to an overblown media sensation and the development of a “myth” [36]. However, our analysis of the actual trial transcripts [42], as below, show that such claims of “myth” do not hold up under scrutiny. Taken as a whole, the recorded testimony and closing arguments demonstrate that the words candy, cola, Twinkies, cupcakes, potato chips, junk food, sugar, or related variants such as brand name soft drinks, were used over two dozen times. Trial transcripts quoted below show that defense attorney Douglas Schmidt leads the defense expert witness, Dr. Martin Blinder, beyond association and into causation:
Defense Attorney: Doctor, you have mentioned this ingestion of sugar and sweets and that sort of thing…does that have any significance, or could it possibly have any significance?
Dr. Blinder: Well, I think, Mr. Schmidt, there are probably three factors that are significant. First, there is a substantial body of evidence that in susceptible individuals large quantities of what we call junk food, high sugar content food with lots of preservatives, can precipitate anti-social and even violent behavior. There have been some studies, for example, where they have taken so-called career criminals and taken them off all their junk food and put them on milk and meat and potatoes, and their criminal records immediately evaporate. There have been a lot of studies in which individuals who are susceptible to these noxious stimuli, when given these noxious stimuli will undergo complete change and engage in behavior which they normally would not. That’s number one.
The crucial part in relation to the Twinkie defense is that Blinder places the ingestion of junk food, which he had just referred to as “noxious stimuli”, into the causative mix:
Dr. Blinder: If it were not for all the tremendous pressures on him the weeks prior to the shooting, and perhaps if it were not for the ingestion of this aggravating factor, this junk food, with all three factors, did not impinge upon him at the same time, I would suspect that these homicides would not have taken place.
Obviously, Dr. Blinder referred to junk food ingestion as the aggravating factor. His reference to a “substantial” body of evidence demonstrating behavioral changes in association with junk food consumption—acting in ways and doing things that are out of the ordinary—provides an inference of a scientific discipline, a specialty. At the time, there was a peppering of historical (low-quality) research indicating that hypoglycemia induced by unhealthy dietary choices could influence EEG results and other neurolaw-related markers [11,43]. Critically, Dr. Blinder underscores that it had been shown that abnormal behavior, even for career criminals, can be remediated by a return to what a reasonable person would assume to be a diet of minimally processed foods. Thus, we have an expert who is informing the jury that science has shown that a diet is to aggression, what smoking is to cancer—an aggravating factor of potential causation.
It is likely that the jury absorbed that messaging because courtroom media picked up on it in real time; following Blinder’s testimony and before jury deliberations had even begun, the Chicago Tribune reported in a nationally-syndicated article that the defense “has been so short on explanations [for diminished capacity] that it seems to have chosen, as one possible rationale, White’s junk food diet”, and the reporting added that the defense psychiatrist was convinced that White’s “deep depressions were escalated by the sugar-heavy diet” [44]. The key word is escalated, which clearly indicates that independent observers were interpreting elements of aggravation and causation. After hearing Dr. Blinder’s testimony, journalist Paul Krassner jotted down the words “Twinkie Defense” and placed them in his real-time reporting for the San Fransisco Bay Guardian [45].
There is additional evidence that White’s claimed junk food diet was being broadly interpreted as a causative factor—the reaction by the prosecution. In Assistant District Attorney (ADA) Thomas Norman’s rebuttal, the prosecution brought in Dr. Roland Levy as their own expert witness. Levy was led into questioning designed to refute the causal diet–crime connections:
ADA Norman: Doctor Levy, are you familiar with any studies and any prevailing scientific bodies of thought relating to the ingestion of sugar, foods with preservatives such as what’s commonly known as junk foods and including, for example, chocolate cupcakes of Twinkie variety, Coca-Cola, candy bars and potato chips, for example, as those relate to being causative factors in influencing anti-social or sociopathic behavior?
Dr. Levy: I am unaware of any prevailing psychiatric opinion that such factors are significant in relationship to any type of mental illness. And I am unaware of any publications in major journals which state that.
Dr. Levy is then cross-examined by White’s defense, chipping at his credibility to discuss nutrition and violence.
Defense: Doctor, I think you testified this morning that you were unaware of any prevailing attitudes or opinions in psychiatry with regard to the ingestion of sugar, or that type of food, is that fair?
Dr. Levy: Yes.
Defense: Would you agree there has been research in that area?
Dr. Levy: There has been.
Defense: There has been much research and publications in that area?
Dr. Levy: Publications, yes.
Defense: And, of course, you have no training or experience with regard to that particular facet of the field?
Dr. Levy: I have never gone into that in any extent at all.
These exchanges clearly demonstrate that the prosecution was well aware of the defense tactics of foods as “causative factors”, and the defense was undermining the expertise of the sole prosecution witness. The problem with Dr. Levy’s response, at least for the prosecution, is that it left the reasonable person with the idea that although connections between highly processed food and violence may not be part of “prevailing” and “major” psychiatric thinking, it could still be part of lesser-known science.
Defense attorney Schmidt opened the door to this novel research in his closing arguments. Schmidt made it clear that even though he could not say definitively that high sugar foods precipitate violence, and that the defense psychiatrist “didn’t go on and on about ‘eat a Twinkie, and go crazy’”, the defense expert had provided important testimony that supports the idea of junk food as an aggravating factor [46]. In other words, one Twinkie was not the culprit, it was a pattern of consumption that intersects with and aggravates mental health vulnerabilities. In his closing arguments, Schmidt revealed his own intentions as to why he called Dr. Blinder: “I don’t know whether there is a relationship between sugar and violent behavior. I don’t know. A lot of people think so. It’s hypoglycemia. If there is some possible relevance regarding that then I want somebody to come in and tell me what it is. And that’s why I called Dr. Blinder”. Schmidt openly acknowledges that Dr. Blinder was brought in to discuss the science of food and antisocial behavior. Schmidt tells the jury that the expert witness for the prosecution, Dr. Levy, essentially called the theory “nonsense” because he was ignorant of the science, “and if I [referring to Dr. Levy with sarcasm] don’t know about it then it’s nonsense”. Schmidt then goes further with the inferences and refers to science outside mainstream dogma: “Whether or not ingestion of foodstuff with preservatives and sugar in high content causes you to alter your personality somehow, or causes you to act in an aggressive manner, I don’t know…there is a minority opinion in psychiatric fields that there is some connection and there very well may be. And if there is a connection, we are working on a reasonable doubt system here. If you have some reason to doubt something, it’s got to be brought up to you because that’s a possible reason for doubting that this was a cold-blooded premeditated murder” [47].

3. Neuroscientist and Jury Reactions

Public reaction to the Twinkie Defense and the trial outcome was not one of support. In the immediate aftermath, California and other states moved swiftly to reduce or eliminate opportunities for diminished capacity as a defense. An article in the San Franscico Examiner noted that “the cream-filled rolls [Twinkies] came to symbolize the public’s disenchantment” with neuropsychiatric testimony [48].
Commenting on the trial in the American Bar Association’s journal Litigation, neurologist Dr. Harold L. Klawans concluded that the defense presentation of White as someone with depression would not have been enough—it was Dr. Blinder’s diet-causation theory that sold the jury [49]. Newsweek magazine’s conclusion was that Dr. Blinder essentially informed the jury “that White’s compulsive diet of candy bars, cupcakes, and Cokes was evidence of a deep depression-and a source of excessive sugar that had aggravated a chemical imbalance in his brain” [50].
While detailed jury-derived explanations for the verdict are lacking, at least one juror stated, “the psychiatric testimony was highly influential. It was mainly what we based the verdict on… it sounded like Dan White had hypoglycemia”, another said, “some people believed it [junk food as a key factor]”, and another added, “I don’t know that much about it [food and behavior]. It’s new research” [51]. From these limited public statements, we know that at least one juror was listening to the defense’s pitch that hypoglycemia can make humans behave abnormally; some on the jury believed the defense propositions, and the latter juror absorbed Blinder’s pitch that the topic was, if nothing else, an emerging science.
In sum, the Twinkie Defense is not a “myth”. There is ample evidence that the defense presented the jury with a scenario in which the defendant’s mental state was aggravated by a reliance upon junk foods, and that aggravation, in the words of the defense attorney in closing arguments, caused the pot “to boil over”. There is also clear evidence that the presentation of the defense did not occur in a vacuum: in the late 1970s in San Francisco, the idea that nutrition could influence behavior was within the cultural petri dish. If there is anything mythical about the Twinkie defense, it is the notion that Twinkies, per se, were presented as a singular item of causation. They were not. The food-like product was simply a culturally relevant symbol of what are now referred to, in the scientific literature, as ultra-processed foods. While we have taken the opportunity in the current essay to provide corrective history, our larger aim is to consider whether or not, four decades later, advances in science lend credibility to the essence of the defense.

4. From Pseudoscience to Contemporary Science

In his critical article in Litigation, neurologist Klawans argued that the prosecution in the White case should have eviscerated the idea that highly processed foods were an aggravating factor: “the Twinkie defense would have been easy to demolish. Mere child’s play. It is not the product of science… There are no data. There never have been. What passes for data are anecdotes and apocrypha, not controlled scientific studies” [49]. While that may have been true in 1979, emergent research from various branches of science supports the idea that dietary patterns, and even the more short-term consumption of macronutrients and dietary additives, can influence brain and behavior [27]. As discussed below, bottom-up research (e.g., microbiome and preclinical) combined with top-down (e.g., nutritional epidemiology) research has been amplified by a collection of recent intervention studies showing that diet is important in behavioral outcomes. Emergent research includes an essential ingredient that was missing from the Twinkie Defense—plausible and reproducible biophysiological mechanisms providing explanations for links between diet and aggression. This includes brain imaging studies showing that the dietary manipulation of gut microbes has the potential to alter the activity of brain regions that control the central processing of emotion and sensation [52].
Before discussing critical findings in the realm, it is worth pointing out that the development of the academic NOVA Food Classification system [53], with its categorical delineation of ultra-processed foods, has allowed international researchers to make a firm case that such foods are detrimental to health, including mental health [54,55]. Ultra-processed items are foods and beverages that typically contain high amounts of refined sugar and/or fats and/or sodium, contain low amounts of naturally occurring dietary fiber and phytochemicals (as found within the minimally processed food matrix), and are typically inclusive of additives such as emulsifiers, flavor enhancers, colors, and isolated fiber and antioxidants that have been removed from their original food matrix; commercially available foods that contain five or more ingredients are generally in the ultra-processed category [56]. Compared to minimally processed foods, ultra-processed foods are also notable for what they typically do not contain—polyphenols and live, non-pathogenic microbes with potential benefits to mental health [57,58]. Dietary patterns high in ultra-processed foods (and/or high fat/sugar) have recently been linked to mental distress [59], various mental disorders [60,61,62,63,64,65,66,67], impulsivity [68,69], diminished concern for future consequences [70], and antisocial/aggressive behavior [71,72,73,74,75,76,77]. Studies using neuroimaging have shown that unhealthy dietary patterns are associated with smaller hippocampal and brain volumes [78,79]. Remarkably, there are indications from human research that even short-term (four day) diets high in added sugars and saturated fat can have a detrimental influence on hippocampal-dependent learning and memory [80].
Despite the large scale of many of the epidemiological studies, they do not provide evidence of causation. However, at least one recent study with a tightly-controlled design (matching macronutrients and total calories in an institutional setting) has shown that ultra-processed foods differ from their minimally-processed counterparts in that they encourage greater caloric consumption and weight gain [81], and produce differences in the human metabolome [82]. In a throwback to White, researchers are demonstrating that vulnerable individuals have the potential to become addicted to ultra-processed foods [83,84]. In the context of mechanisms described below, ultra-processed food consumption has recently been associated with low-grade systemic inflammation [85,86,87] and appears to influence systemic metabolites and energy uptake via disturbances to the gut microbiome [88,89,90,91]. In recent years, scientists have also developed a more sophisticated understanding of the ways in which specific nutrients, ranging from vitamins and minerals to omega-3 fatty acids, influence the structure and function of the brain [92]. Moreover, dietary fiber and naturally occurring phytochemicals (e.g., polyphenols and related chemicals that give food their colors, taste, and texture), constituents of minimally-processed foods, are also involved in cognition and behavior via direct and indirect effects on nervous system health [93].

5. Mechanistic Pathways

There are a number of ways in which dietary patterns and individual dietary components can influence (i.e., promote the integrity of, or compromise) brain structure and function. Either directly, or through metabolic reactions, multiple nutrients (and non-nutritive phytochemicals such as polyphenols) play critical roles in maintaining the integrity of neuronal membranes, neurotransmitter production, neurogenesis, and protection through antioxidant defense systems [94,95]. Ultra-processed foods and the synthetic additives within can compromise dopamine and serotonin neurotransmission, through influence on gene transcription and, as discussed below, inflammation [96,97]. Preclinical studies show that these foods and the additives within can disrupt the amygdala–hippocampal complex, which, extrapolated to humans, suggests an influence on the frontolimbic emotion regulation brain network; indeed, ultra-processed food consumption in humans has been associated with lower volumes in mesocorticolimbic areas of the brain [98].
In the context of healthy dietary patterns and the brain, there is significant nutritional complexity in structure–function connections; that is, single nutrients often work in relation to others. For example, the omega-3 fatty acids docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA) play important roles in neuronal structure and signaling; however, these fatty acids are dependent on B vitamins for transport [99]. Another example is magnesium, which has been linked to antisocial behavior [100]. It is difficult to pinpoint precise mechanisms because magnesium plays a role in hundreds of enzymatic reactions and interacts along with other nutrients. Pertinent to our discussions of dietary glutamate below, low levels of magnesium exacerbate neuronal excitotoxity [101], while omega-3 fatty acids protect against monosodium glutamate neurotoxicity [102]. There is also evidence that chronic psychological stress increases biophysiological demand for a variety of micronutrients [103]. This might be especially the case with nutrients involved in the antioxidant defense system and those involved in immune function, including zinc, magnesium, iron, and various B vitamins [103].
One mechanistic area that has received considerable attention is the role of dietary patterns and individual dietary components on neuroinflammation. Volumes of research now point to inflammation as a causative factor in neuropsychiatric conditions. Included in this research are studies linking elevations in immune proteins (e.g., cytokines) in adults with aggressive behavior and/or aggressive tendencies [104,105,106,107]. Both pre-clinical and human research indicates that chronic microglia activation in the brain may contribute to various neuropsychiatric disorders, a loss of impulse control, addiction, and aggressive behavior [108,109,110]. Neuroinflammation is related to excitotoxity (the overactivity of neurons) and oxidative stress within the central nervous system. Together, neuroinflammation, oxidative stress, and excitotoxicity, are known as the ‘neurotoxic triad’ and each component can influence the other in a potentially self-sustaining and synergistic manner [111]. Conditions for the neurotoxic triad are made worse by psychological trauma and acute and chronic stress which compromise the structure and function of the normal blood–brain barrier (BBB). This allows the infiltration of chemicals normally excluded to the periphery, including inflammatory cytokines, to gain central access [112]. Diet plays a crucial role in the neurotoxic triad because certain dietary patterns, such as those rich in vitamins, minerals, fiber, omega-3 fatty acids, and phytochemicals, have been shown to limit neuroinflammation [111]. On the other hand, patterns rich in high-sugar/high-fat ultra-processed foods, often containing various emulsifiers and flavor enhancers (e.g., free-form glutamate, monosodium glutamate (MSG), and aspartame), have been linked with the neurotoxic triad [113].
Animal research using germ-free models indicates that stress can directly elevate systemic inflammation in the absence of microbiota [114]. However, a growing body of international research demonstrates that the gut microbiome plays an important mediating role in the links between dietary patterns (and specific components of diet) and neuroinflammation [115]. As discussed in the next section, emerging research suggests that the dysbiosis (disturbances to the microbiome) observed in most neuropsychiatric conditions is not merely an association or consequence of a disease or disorder [116].

6. Microbiome and the Legalome

Although much of the human research linking microbial alterations with emotional regulation and neuropsychiatric disorders is cross-sectional [117], researchers are moving beyond mere associations [118]. Dietary patterns high in ultra-processed foods (vs. minimally processed, polyphenol, and fiber-rich diets) have been linked to gut dysbiosis [91,119,120,121]. Individual components of ultra-processed foods, such as synthetic emulsifiers and dietary excitotoxins, have been linked to dysbiosis [122,123]. On the other hand, human research shows that targeting the microbiome via diet (e.g., whole grains, onions, leeks, cabbage, oats, and fermented foods) can reduce psychological stress and improve mood [124,125].
The ultra-processed food induction of gut dysbiosis is important because preclinical studies are indicating that gut microbes play a causative role in cognitive–behavioral disturbances, including aggression and antisocial behavior. For example, when the fecal material of animals with diet-induced dysbiosis is transplanted into otherwise healthy animals, these recipients have observable behavioral disturbances as found in the dysbiotic donors [126,127,128]. These behavioral signals via microbiota transfers have also been observed when the fecal material originated from human donors with behavioral disorders [129,130,131,132]. When animals are the recipients of fecal material obtained from human donors with mild cognitive impairment, recipients experience learning and memory problems in association with impaired cerebral glucose uptake [133]. Of relevance to addiction, microbiota from alcohol-dependent patients induced the behavioral alterations associated with alcohol dependence in recipient lab animals, including increased anxiety- and depression-like behaviors, reduced exploratory and recognition memory, and higher alcohol preference; these behavioral changes were accompanied by objective brain-related signals known to be associated with alcohol dependence [134]. Remarkably, the transfer of fecal material from human infants with microbial disruptions (from the administration of antibiotics) leads to aggressive-like behavior in recipient lab animals, observations not seen with transfer of microbiota from healthy infants [135]. When combined with other preclinical fecal transfer studies, the emerging picture is that the human equivalents of depression, anxiety, and behavioral disorders are (at least to some degree) rooted in, and transmissible by, gut microbes [136].
The mechanisms explaining such observations are beginning to take shape; in particular, gut dysbiosis leads to intestinal permeability (a more porous intestinal lining, or so-called ‘leaky gut), increased levels of circulating immune chemicals, most notably proinflammatory cytokine responses, and an ongoing low-grade inflammation [137,138]. This low-grade inflammation is considered a risk factor for a variety of neuropsychiatric conditions [107,139]. In animals, exposure to ultra-processed diets can increase the permeability of both the gut and the aforementioned BBB [140]. This low-grade inflammatory cascade and associated metabolic dysregulation can influence mood and aggression [141,142]. Intestinal permeability allows gut microbial breakdown products, such as lipopolysaccharide endotoxin (LPS), to enter circulation. LPS provokes inflammatory cytokine release and enhances the potential of dietary excitotoxins such as MSG to promote neuroinflammation and the dysfunction of neurotransmission [143]. Animal studies show that MSG can influence serotonin, dopamine, and norepinephrine levels [144,145], and alter the gut microbiome [146]. The formation of bioactive polyphenolic metabolites via microbial activity in the gut lumen, with preferred delivery to the mammalian brain [147], appears to play an important mechanistic role in mental and cognitive health [93].
Since research is pointing toward a causal role for gut microbiota in systemic inflammation [148], scientists are actively pursuing gut microbial signatures that can tie together intestinal permeability, systemic low-grade inflammation, and the risk of aggression [149,150]. It is also true that alterations in the gut microbiome and the cascade of systemic low-grade inflammation can be mediated from the top-down, i.e., stress activates brain to gut pathways, altering the gut microbiome [151]; these stress-induced changes to the microbiome appear to drive and maintain psychological symptoms via microbial influences on various metabolic pathways [152]. In human research, reports point toward linkages between select microbes that appear to be involved in reactive aggression, such as Lachnospiraceae and Eubacterium genera, that thrive with unhealthy dietary patterns absent in phytochemical-rich vegetables [153].
The rapidly emerging research in microbiome sciences has already intersected with criminal courts and considerations of culpability. As described in detail elsewhere, cases of forensically relevant alcohol production from the human gastrointestinal tract (i.e., auto-brewery syndrome), without any consumption of alcohol-containing beverages/foods, are increasingly described in the medical literature [18]. This internal alcohol production, which can bring a person close to or above legal limits of blood alcohol, is a product of dysbiosis, most notably the overgrowth of yeasts such as Candida and bacterium such as Klebsiella [154]. Since the acute microbially driven alcohol production is via high-sugar, ultra-processed drinks/foods (with dysbiotic microbes acting on ingested sugar to produce alcohol), the resultant inebriated state and “brain fog” raises questions of diminished capacity and criminal intent. In both the United States and Europe, cases of DWI have recently been dismissed due to lab-proven internal alcohol production (i.e., with zero consumption of any alcohol containing product) [18] (Figure 1). It is important to note that blood alcohol levels far below DWI-related legal standards are associated with cognitive changes and a higher risk of injury [155]. The recent court rulings are, at least to some degree, a validation of the ideas first floated around the time of The People v. White, and indicate that the courts are willing to accept lab-proven connections between diet, gut microbes, and the production of metabolites that can otherwise influence cognition and behavior. As noted in a recent editorial in Trends in Microbiology, the available microbiota–gut–brain axis research is already forcing important policy questions upon society, including efforts to remedy systemic health inequalities [156]. We suggest that the legalome (microbiome and omics science applied in forensic and legal psychology) will be part of those salient policy discussions.

7. Intervention Studies

The case for claimed causality is strengthened by emerging nutritional intervention studies. In the immediate aftermath of The People v. White, some researchers began examining the relationships between diet and antisocial behavior in correctional settings. In a series of quasi-experimental studies across twelve different US juvenile correctional institutions, investigators swapped out foods with high amounts of added sugar and refined fats for similar, less processed options [157,158,159,160]. Combined, the studies involved several thousand juveniles with the results showing an average 47% reduction in documented offenses, infractions, and other indicators of antisocial behavior. These included reductions in overt violence, acts of theft, verbal aggression, and insubordination to corrections personnel [161,162].
While encouraging, these dietary interventions studies from the 1980s lacked tightly controlled comparison groups and double-blind design, and undoubtedly suffered from expectancy effects among participants. With a better understanding of potential mechanisms, contemporary researchers have revisited the topic of nutritional neuropsychiatry with more tightly controlled research designs. Many of the emerging nutritional neuroscience/psychology studies examine depression as an outcome. In the context of forensic psychology, the positive influence of diet on depression is important because research has shown that depressive symptoms are associated with cognitive disturbances related to legal capacity [163,164,165,166,167,168], enhanced risk-taking [169], and deficits in social problem-solving [170]. The SMILES trial (n = 67) found improved ratings of depression on a clinical rating scale with a healthy diet intervention compared to a social support control group [171]. It appears that the positive results of the SMILES trial were related to the elimination of ultra-processed foods, rather than the introduction of specific healthy foods [172]. Research suggests that mental health can be influenced by relatively short-term dietary changes. For example, in a randomized dietary intervention study involving young adults (n = 101), a switch to a healthy dietary pattern (vs. habitual diet) lowered the symptoms of depression in three weeks; the significant improvements were also documented at a 3 month follow-up [173]. In a multi-center, randomized controlled trial (n = 292), researchers reported that a low-fat, low-glycemic index, plant-based diet intervention improved mental outlook and work productivity in adults [174]. These studies are joined by similarly-designed randomized controlled intervention studies, demonstrating improved mental health with adherence to dietary patterns emphasizing fruits, vegetables, lean meats, fish, and whole grains, at the exclusion of highly processed snacks and fast food [171,175,176,177,178,179].
In an older non-randomized intervention trial, the addition of citrus juice in a juvenile correctional facility was associated with a 47% reduction in antisocial behavior [180]. More recent research shows that the oral intake of juices rich in naturally occurring flavonoids improves mood in young adults with depressive symptoms [181], and lower scores on anger/hostility measures in otherwise healthy adults have also been noted [182]. In addition, human studies have also evaluated the response to the removal/limitation of components commonly found in ultra-processed foods. Removing dietary excitotoxins (e.g., aforementioned free-form glutamates, MSG, and aspartame) shows benefit along a number of neuropsychiatric lines [183,184]. The metagenomic analysis of fecal material is allowing researchers to evaluate the accuracy of self-reported food intake among research participants, advances that will undoubtedly strengthen the validity of outcomes in future studies [185].
Although our focus here has been on dietary patterns, rather than isolated nutrients, it is worth pointing out that some of the important constituents of healthy dietary patterns, such as omega-3 fatty acids, may play a role in curbing aggression and antisocial behavior. For example, a recent meta-analysis of 28 randomized-controlled trials (n = 3918) concludes that omega-3 fatty acids modestly, but significantly, reduce aggression in both children and adults [186]. The study of dietary supplements (e.g., multivitamin–mineral formulas, zinc, magnesium, vitamin D, and food-derived polyphenols) for the reduction of aggression and antisocial acts (especially in correctional settings) remains limited, although encouraging results have been noted [187,188]. In addition to the direct influence of omega-3 fatty acids and other individual nutrients on neurotransmission, and inflammation-lowering effects, beneficial outcomes may be mediated by influence on the gut microbiome [189]. Related to this are the growing number of human studies indicating that ‘psychobiotic’ agents (e.g., probiotics and prebiotics) can influence social decision-making [190], buffer stress and improve mood [191,192], and lower aggressive thoughts [193], aggressive actions [194], and impulsivity [195].

8. Where to Next?

In order for nutritional neuroscience to have increased application in the context of neurolaw, many questions will need to be answered, especially those that close the gap between correlation and causation. Even though there is an increasing number of controlled intervention studies showing that healthy dietary patterns (and the avoidance of ultra-processed foods) can influence mood and cognition, these have typically not considered justice-involved people. Future studies should include justice-involved populations, pairing controlled dietary interventions with behavioral outcomes and objective markers, including gut microbiome indicators, inflammatory cytokines, indicators of leaky gut, and those drawn from omics technologies (including genomics, epigenomics, metabolomics, and/or transcriptomics). Given the emerging research on the gut microbiome and temperament [196], there is a need to study the microbiome in justice-involved populations [197], while examining potential dietary links and the neurobehavioral consequences of dysbiosis [198].
The area of ultra-processed food addiction and its potential relationship to antisocial behavior is worth pursuit. Accumulating human and preclinical research show that ultra-processed foods have addictive properties, and attempts to stop consuming such products induce withdrawal-like signs and symptoms [199]. Neuroimaging studies suggest there may be common pathways underpinning the observation of reward–behavior in individuals engaging in antisocial activity [200]. Recent evidence suggests that the experience of victimization (e.g., through violence or bullying) increases the likelihood of ultra-processed food consumption [201]. Since many offenders have a prior history of being victimized, this relationship is worthy of analysis. To what extent is the chronic consumption of hyperpalatable foods an attempt at reducing stress [202]? Ultra-processed food consumption has also been linked to other known risk factors for antisocial behavior, including alcohol consumption and illicit drug use [203]. The microbiome may provide useful markers of vulnerability to ultra-processed food addiction, and guide intervention efforts [204]. Clinicians and researchers now have validated tools which can be used to screen for the symptoms of ultra-processed food addiction [205].
Another area worth pursuit is whether or not ultra-processed foods can influence neurocognition above and beyond any outcomes that can be traced to macronutrients such as high sugar content. Although ultra-processed foods can contain high amounts of sugar, the NOVA classification system is not ideal for separating out high-glycemic index foods [206]. Consumers can wade through foods that are technically ultra-processed and still find items that are low in sugar and inclusive of significant levels of nutrients. On the other hand, relying only on the sugar or fat content of a food or beverage can overlook the ways in which an artificial sweetener (e.g., aspartame) or flavor enhancer (e.g., MSG and its many variants) could influence neurocognitive outcomes. One way to examine this would be to study the effects of different meals (with manipulated levels of processing/additives) in laboratory designs that are intended to (potentially) lead to signs of aggression and antisocial behavior. Simulated long-distance urban driving, intended to assess fatigue and ‘road rage,’ is an example where post-prandial effects could be studied. Research already indicates that aggression may be a common response to reactive hypoglycemia [207,208]; however, it is unknown whether aggression and antisocial behavior might differ when lab-induced self-control fatigue is examined when participants first consume meals differing in processing levels, sugar content, and excitotoxin additives.
The study of nutritional neuroscience as it intersects with the courts has the potential to illuminate some of the vague diagnostic conundrums common to clinical medicine in general, and neurologists in particular. For example, “brain fog” is a term often used by patients as a way to express a long continuum of a lack of attentional focus and cognitive fatigue [209]. Brain fog can be a descriptive for the depersonalization and derealization that is associated with some criminal acts [210,211,212]. Recently, validated instruments have emerged to assess brain fog [213]. Brain fog is common to individuals with gastrointestinal complaints, and brain fog questionnaires are capable of distinguishing between established measures of obvious cognitive impairment (e.g., the Montreal Cognitive Assessment and the Digital Symbol Substitution test) and the mental fatigue, exhaustion, drowsiness, and difficulty thinking and focusing that otherwise characterizes brain fog [214]. These brain fog assessments can be paired with objective neuroimaging, EEG testing, biological samples (omics-related), and mobile technology-assisted ‘ecological momentary assessment’ as a way to better understand how diet might intersect with cognition and behavior. How might a concept such as brain fog relate to aggression and unplanned criminal activity (so-called irresistible impulse), and what are its biological underpinnings? Since brain fog has been associated with dysbiosis and an elevated production of D-lactic acid [215,216], and elevated lactic acid, especially D-lactate, has been linked to anxiety, aggression, and signs/symptoms that mimic alcohol inebriation [217,218,219,220,221], once again, microbiome insights might be helpful. There is considerable variation in plasma D-lactate levels among the general population, and it is worth noting that the microbial species often linked to auto-brewery syndrome, Klebsiella pneumoniae [18], is also efficient at converting glucose into D-lactate [222]. The growth of Klebsiella pneumoniae is promoted by dietary patterns dominated by sugar and simple carbohydrates, with absent naturally occurring fiber [223].
In relation to the Twinkie Defense, and the legalome in forensic psychology, the question is how might the absence of these nutrients (omega-3 fatty acids, polyphenols, and vitamins/minerals) in an ultra-processed food-heavy diet [57,224,225], and the presence of mood-altering additives (e.g., glutamate flavor enhancers) [183,226] influence diminished capacity and intent? How does the intake of one nutrient (e.g., vitamin D) offset the potential adverse cognitive–behavioral consequences of flavor enhancers and other synthetic food additives [227]? How do these potentially harmful ingredients add to the addictive properties of ultra-processed foods [228], and how does the hyper-palatability [229] influence a vulnerable person toward ‘comfort food’ as a means to palliate isolation and depression, as claimed by the defense in The People v White. There is also a need to better understand the frequency of auto-brewery syndrome and how ultra-processed foods and dysbiosis might combine to produce alcohol and a host of other chemicals with forensic implications [18]. How could advances in breath tests (e.g., those that measure fasting and post-prandial metabolites) help researchers with better diagnostics that might be used in risk assessments?
The subject of dietary tryptophan (as a precursor of serotonin) is also of importance to the relationship between ultra-processed foods and antisocial behavior. Preclinical studies show that high fat/high sugar diets can alter serotonergic gene expression, likely mediated by the microbiome [230]. Recent human studies have linked tryptophan-rich diets with better mental health and social cognition [231,232]. Dietary tryptophan appears to enhance emotion recognition, affiliation with victims, and cognitive empathy [233]. In addition, multiple experimental studies have linked dietary tryptophan depletion with increased aggression, quarrelsome behavior, and diminished agreeableness [234]. Ultra-processed foods in the Nova 4 category are typically not rich in tryptophan [235]. Given the widespread use of ultra-processed foods in carceral systems, how might the tryptophan content of the diet relate to outcomes ranging from rule infractions to acts of major violence in prisons? How might gut microbes, which produce various tryptophan metabolites, including those with neuroactive properties, factor into the links between dietary tryptophan and behavior [236]?
Finally, the defense in The People v. White referred to the phenomenon of hypoglycemia as a possible explanation for violence. At the time of trial, the notion that low blood sugar, following the consumption of high-sugar, low-protein meals, is a contributor to neuropsychiatric complaints and aggressive behavior was supported by limited evidence [237,238]. In the years immediately after the trial, small-scale laboratory studies linked low blood glucose with aggressive reactions (measured after an oral glucose beverage), in both women and men [239,240]. Importantly, these associations were not dependent upon very low levels of blood glucose that typify diagnostic hypoglycemia. Researchers also reported that among criminal offenders (vs. healthy non-offender controls), an oral glucose challenge leads to initially higher levels of blood glucose and a subsequent lower glucose nadir in the period following glucose consumption [241,242,243,244]. More recent evidence supports the idea that, after initial consumption, glucose reduces aggressive tendencies and increases helping behavior under stress [245,246]. However, this benefit may be short-lived. Low blood glucose is associated with aggression in healthy adults [207,247]. Is it possible that a vulnerable person might resort to aggression in the period following the consumption of a high-sugar ultra-processed food, as blood glucose levels fall? Initial observations linking violence with both low blood glucose (after glucose challenge) and deficits in central neurotransmission [248] are worthy of further investigation.

9. Conclusions

Although the roots of neurolaw and the use of objective measurements (e.g., EEG) in the courts date back almost a century, recent years have witnessed an increased uptake of neuroscience findings in criminal and civil courts. Discoveries in the neurosciences and related interdisciplinary fields, especially those that illuminate structure/function relationships to behavior, are actively challenging traditional legal assumptions surrounding blame, responsibility, criminal intent, agency, and moral responsibility [249]. While neurolaw does not challenge society’s right to protect itself, and to separate individuals that present a danger, it does confront matters of punishment and notions of equally distributed free will [250]. Among the exciting advances in neuroscience-related research, the role of nutrition and synthetic additives in nervous system structure and function has received increased attention. In addition, microbiome studies, especially preclinical work involving fecal transplants, are indicating that gut microbes make a significant contribution to mammalian cognition and behavior. Since the microbiome is dependent upon diet and other environmental factors, it is no longer possible to dismiss nutrition as an important factor in neurocognition [251].
In forensic neuropsychology texts and the popular press, the Twinkie Defense is often claimed to be a ‘myth’. However, using court transcripts we have shown that discussions of ultra-processed foods as a causative factor were suggested in testimony and in closing arguments, and attempts were made by the prosecution to refute causal connections. The case had a chilling effect on the defense of diminished capacity, depreciated trust in neuropsychiatric testimony, and likely minimized the seriousness of diet–brain–behavior interactions [252]. Still, even in the 1980s, the Twinkie Defense was credited with accelerating robust debate on the role of neuroscience in matters of criminal responsibility and the morality of punishing those with neuropsychiatric disorders [1]. Forty-five years later, emerging research in neurolaw and the study of the biological underpinnings of violence, aggression, antisocial behavior, and criminal behavior writ-large, underscore the terms intersectionality and vulnerability [14]. Based on genetics, epigenetics, lived experiences (and all of the associated adverse exposures or positive assets), and innumerable inputs across the spectrum of biopsychosocial factors (especially those related to antisocial behavior and justice involvement [253]), how might dietary inputs intersect with the vulnerabilities of an individual [254]? Based on the available evidence in the realm of nutritional criminology, we suspect that dietary inputs contribute to (and press upon existing) vulnerabilities of criminal behavior in ways that are more consequential than currently appreciated in the criminal justice system [26].
The American Psychological Association’s (APA) recent inclusion of Continuing Education (CE) modules on nutrition and behavior in its flagship Monitor on Psychology, has sent a signal that the topic of nutrition and behavior is here to stay. It is noteworthy that the lead article introducing the APA CE series acknowledged that researchers in nutritional neuroscience have historically “encountered skepticism and dismissiveness from many health care professionals and researchers, including psychologists, who tend to downplay the importance of diet in health and mental health” [255]. Based on increasingly robust mechanistic evidence, that dismissiveness is changing. Although the APA CE did not venture into the criminal justice system applications, the pace of nutritional neuroscience and attention to neurolaw, indicates that such applications are an inevitability. Although many unanswered questions remain, if the research continues to strengthen on its current trajectory, experts in forensic neurosciences can expect to be called upon with increased frequency, especially at the intersection of nutrition and diminished capacity. In the meantime, it is essential that critically relevant findings in neuroscience permeate criminal justice discourse.

Author Contributions

Conceptualization, S.L.P. and A.C.L.; review and editing, K.F.H., C.A.L., and J.J.N.; investigation and formal analysis of court transcripts, A.C.L.; writing—original draft preparation, S.L.P. and A.C.L.; supervision, K.F.H. and C.A.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

C.A.L. is Cofounder, Board Member, and Chief Scientific Officer of Mycobacteria Therapeutics Corporation, and is a member of the faculty of Clinical Care Options, LLC (CCO), Reston, Virginia, the Integrative Psychiatry Institute, Boulder, Colorado, the Institute for Brain Potential, Los Banos, California, and Intelligent Health Ltd., Reading, UK. In the previous three years, C.A.L. served on the Scientific Advisory Board of Immodulon Therapeutics Ltd., London, UK. All other authors declare no conflicts of interest.

References

  1. Makin, K. Is crime the product of an abnormal mind? Medical evidence now suggests that many offenders don’t really control the things they do. The Globe and Mail, 21 January 1988; A-7. [Google Scholar]
  2. Jones, O.D.; Wagner, A.D.; Faigman, D.L.; Raichle, M.E. Neuroscientists in court. Nat. Rev. Neurosci. 2013, 14, 730–736. [Google Scholar] [CrossRef] [PubMed]
  3. Canela, C.; Buadze, A.; Dube, A.; Jackowski, C.; Pude, I.; Nellen, R.; Signorini, P.; Liebrenz, M. How do legal experts cope with medical reports and forensic evidence? The experiences, perceptions, and narratives of swiss judges and other legal experts. Front. Psychiatry 2019, 10, 18. [Google Scholar] [CrossRef] [PubMed]
  4. Giannouli, V.; Tsolaki, M. Legal capacity of the elderly in Greece. Hell. J. Nucl. Med. 2014, 17, 2–6. [Google Scholar] [PubMed]
  5. Barsky, B.A.; Stein, M.A. The United Nations convention on the rights of persons with disabilities, neuroscience, and criminal legal capacity. J. Law Biosci. 2023, 10, lsad010. [Google Scholar] [CrossRef]
  6. Schneider, B.S.; Arciniegas, D.B.; Harenski, C.; Clarke, G.J.B.; Kiehl, K.A.; Koenigs, M. The prevalence, characteristics, and psychiatric correlates of traumatic brain injury in incarcerated individuals: An examination in two independent samples. Brain Inj. 2021, 35, 1690–1701. [Google Scholar] [CrossRef] [PubMed]
  7. Witzel, J.G.; Bogerts, B.; Schiltz, K. Increased frequency of brain pathology in inmates of a high-security forensic institution: A qualitative CT and MRI scan study. Eur. Arch. Psychiatry Clin. Neurosci. 2016, 266, 533–541. [Google Scholar] [CrossRef]
  8. Taylor, E.N.; Timko, C.; Nash, A.; Owens, M.D.; Harris, A.H.; Finlay, A.K. Posttraumatic stress disorder and justice involvement among military veterans: A systematic review and meta-analysis. J. Trauma. Stress 2020, 33, 804–812. [Google Scholar] [CrossRef]
  9. Hall, D.; Lee, L.-W.; Manseau, M.W.; Pope, L.; Watson, A.C.; Compton, M.T. Major mental illness as a risk factor for incarceration. Psychiatr. Serv. 2019, 70, 1088–1093. [Google Scholar] [CrossRef]
  10. Hayes, H. Neurolaw: The Intersection of Science and the Law. Perspectives 2017, 25, 12. [Google Scholar]
  11. Logan, A.C.; Nicholson, J.J.; Schoenthaler, S.J.; Prescott, S.L. Neurolaw: Revisiting Huberty v. McDonald’s through the Lens of Nutritional Criminology and Food Crime. Laws 2024, 13, 17. [Google Scholar] [CrossRef]
  12. Fozdar, M.A. The history of forensic neuropsychiatry. Behav. Sci. Law 2024, 42, 20–27. [Google Scholar] [CrossRef] [PubMed]
  13. Sapolsky, R.M. Determined: A Science of Life without Free Will; Penguin Press: London, UK, 2023. [Google Scholar]
  14. Sapolsky, R.M. Behave: The Biology of Humans at our Best and Worst; Penguin Press: London, UK, 2017. [Google Scholar]
  15. Steele, K.A. The law, the science, and the logic of ending the teenage death penalty. J. Pediatr. Neuropsychol. 2021, 7, 9–26. [Google Scholar] [CrossRef]
  16. Catley, P. The Need for a Partial Defence of Diminished Capacity and the Potential Role of the Cognitive Sciences in Helping Frame That Defence. In Neurolaw: Advances in Neuroscience, Justice & Security; Springer: Berlin/Heidelberg, Germany, 2021; pp. 51–75. [Google Scholar]
  17. Beech, A.R.; Carter, A.J.; Mann, R.E.; Rotshtein, P. The Wiley Blackwell Handbook of Forensic Neuroscience, 2 Volume Set; John Wiley & Sons: Hoboken, NJ, USA, 2018; Volume 1. [Google Scholar]
  18. Logan, A.C.; Prescott, S.L.; LaFata, E.M.; Nicholson, J.J.; Lowry, C.A. Beyond Auto-Brewery: Why Dysbiosis and the Legalome Matter to Forensic and Legal Psychology. Laws 2024, 13, 46. [Google Scholar] [CrossRef]
  19. Pollack, H.A.; Dosani, E. The Public Health Community Must Stand up for Greater Re-Sources Devoted to Criminal Justice Research. Milbank Q July 30. 2024. Available online: https://www.milbank.org/quarterly/opinions/the-public-health-community-must-stand-up-for-greater-resources-devoted-to-criminal-justice-research/ (accessed on 1 August 2024).
  20. Taylor, J.; Harp, J.; Elliott, T. Meeting the legal challenge. Neurolaw Lett. 1991, 1, 1. [Google Scholar]
  21. Taylor, J.S.; Harp, J.A.; Elliott, T. Neuropsychologists and neurolawyers. Neuropsychology 1991, 5, 293. [Google Scholar] [CrossRef]
  22. Shen, F.X. The overlooked history of neurolaw. Fordham L. Rev. 2016, 85, 667. [Google Scholar]
  23. Brown, E. Is “Neurolaw” Coming Soon to a Courtroom Near You? Scientific American. 7 September. Available online: https://www.scientificamerican.com/article/is-neurolaw-coming-soon-to-a-courtroom-near-you/ (accessed on 9 September 2024).
  24. Zaalberg, A. The effects of nutrients and neurotoxicants on aggressive behavior. J. Crim. Just. 2019, 65, 101592. [Google Scholar] [CrossRef]
  25. Poulter, M.; Coe, S.; Graham, C.A.-M.; Leach, B.; Tammam, J. A Systematic Review of the effect of Dietary and Nutritional Interventions on the Behaviours and Mental Health of Prisoners. Br. J. Nutr. 2024, 1–32. [Google Scholar] [CrossRef]
  26. Prescott, S.L.; Logan, A.C.; D’Adamo, C.R.; Holton, K.F.; Lowry, C.A.; Marks, J.; Moodie, R.; Polland, B. Nutritional Criminology: Why the Emerging Research on Ultra- Processed Food Matters to Health and Justice. Int. J. Environ. Res. Public Health 2024, 21, 120. [Google Scholar] [CrossRef]
  27. Heidari, M.; Khodadadi Jokar, Y.; Madani, S.; Shahi, S.; Shahi, M.S.; Goli, M. Influence of food type on human psychological—Behavioral responses and crime reduction. Nutrients 2023, 15, 3715. [Google Scholar] [CrossRef]
  28. Tcherni-Buzzeo, M. Dietary interventions, the gut microbiome, and aggressive behavior: Review of research evidence and potential next steps. Aggress. Behav. 2023, 49, 15–32. [Google Scholar] [CrossRef] [PubMed]
  29. Gulledge, L.; Oyebode, D.; Donaldson, J.R. The influence of the microbiome on aggressive behavior: An insight into age-related aggression. FEMS Microbiol. Lett. 2023, 370, fnac114. [Google Scholar] [CrossRef] [PubMed]
  30. Robinson, M. The Food IS the Crime: A Focus on Food as “Food Crime”. Int. J. Crim. Justice Sci. 2022, 17, 167–187. [Google Scholar]
  31. Robinson, M. Eating ourselves to death: How food is a drug and what food abuse costs. Drug Sci. Policy Law. 2022, 8, 20503245221112577. [Google Scholar] [CrossRef]
  32. Torrey, E.F. Freudian Fraud: The Malignant Effect of Freud’s Theory on American Thought and Culture; Harper Collins Publishers: New York, NY, USA, 1992. [Google Scholar]
  33. Lattey, R.M.D. Sigmund Freud, pseudoscientist. Can. Fam. Physician 1969, 15, 59–63. [Google Scholar]
  34. Denno, D.W. Criminal law in a post-Freudian world. Univ. Ill. Law Rev. 2005, 2005, 601. [Google Scholar]
  35. Hasse, A.F. Keeping Wolff from the Door: California’s Diminished Capacity Concept. Calif. L. Rev. 1972, 60, 1641. [Google Scholar] [CrossRef]
  36. Pogash, C. Myth of the ‘Twinkie Defense’/The Verdict in the Dan White Case Wasn’t Based on His Ingestion of Junk Food. San Francisco Chronicle. 23 November. Available online: https://www.sfgate.com/health/article/Myth-of-the-Twinkie-defense-The-verdict-in-2511152.php (accessed on 20 August 2023).
  37. Snider, K. The infamous Twinkie defense--fact or fiction? Ann. Am. Psychother. Assoc. 2006, 9, 42–44. [Google Scholar]
  38. Walker, L.E.; Shapiro, D.; Akl, S.; Walker, L.E.; Shapiro, D.; Akl, S. Criminal Responsibility. In Introduction to Forensic Psychology: Clinical and Social Psychological Perspectives; Walker, L.E., Shapiro, D.A., Eds.; Springer: New York, NY, USA, 2020; pp. 37–51. [Google Scholar]
  39. Ewing, C.P.; McCann, J.T. Minds on Trial: Great Cases in Law and Psychology; Oxford University Press: Oxford, UK, 2006. [Google Scholar]
  40. Halpern, D.F.; Brooks, A.; Stephenson, C. How science by media creates false certainties and resistance to conceptual change. In Malingering, Lies, and Junk Science in the Courtroom; Kitaeff, J., Ed.; Cambria Press: Amherst, NY, USA, 2007; pp. 15–33. [Google Scholar]
  41. Raine, A. The Anatomy of Violence; Pantheon Books: New York, NY, USA, 2013. [Google Scholar]
  42. Linder, D.O. The Trial of Dan White: Trial Testimony of Dr. Martin Blinder (Defense Psychiatrist). Famous Trials. University of Missouri-Kansas City School of Law. Available online: https://www.famous-trials.com/danwhite/601-blindertestimony (accessed on 19 August 2023).
  43. Fishbein, D.; Pease, S.; Pung, O.B. The Effects of Diet on Behavior: Implications for Criminology and Corrections; Robert J. Kutak Foundation: Omaha, NE, USA, 1988. [Google Scholar]
  44. Coakley, M. What turned politician into a killer? Macon Telegraph and News, 13 May 1979; 8-A. [Google Scholar]
  45. Krassner, P. Sex, Drugs, and the Twinkie Murders; Loompanics Unlimited: Port Townsend, WA, USA, 2000. [Google Scholar]
  46. Jennings, D. Dan White jury hears the final arguments. San Franscisco Chronicle, 16 May 1979; 1+14. [Google Scholar]
  47. Salter, K. The Trial of Dan White; Market Systems, Inc.: El Cerrito, CA, USA, 1991; p. 386. [Google Scholar]
  48. Ludlow, L. How White used the ‘Twinkie Defense’ and why it will be hard to use it again. The San Francisco Examiner (San Francisco, California), 9 September 1983; 6. [Google Scholar]
  49. Klawans, H.L. The Twinkie Defense. Litig 1991, 18, 59–64. [Google Scholar]
  50. Beck, M.; Reese, M. Night of gay rage. Newsweek 1979, 93, 30–31. [Google Scholar]
  51. Pogash, C. Inside the White jury room. The San Francisco Examiner (San Francisco, California), 24 May 1979; 1+26. [Google Scholar]
  52. Tillisch, K.; Labus, J.; Kilpatrick, L.; Jiang, Z.; Stains, J.; Ebrat, B.; Guyonnet, D.; Legrain–Raspaud, S.; Trotin, B.; Naliboff, B. Consumption of fermented milk product with probiotic modulates brain activity. Gastroenterology 2013, 144, 1394–1401.e4. [Google Scholar] [CrossRef] [PubMed]
  53. Monteiro, C.A.; Cannon, G. The trouble with ultra-processed foods. BMJ 2022, 378, o1972. [Google Scholar] [CrossRef]
  54. Prescott, S.L.; D’Adamo, C.R.; Holton, K.F.; Ortiz, S.; Overby, N.; Logan, A.C. Beyond Plants: The Ultra-Processing of Global Diets Is Harming the Health of People, Places, and Planet. Int. J. Environ. Res. Public Health 2023, 20, 6461. [Google Scholar] [CrossRef] [PubMed]
  55. Touvier, M.; da Costa Louzada, M.L.; Mozaffarian, D.; Baker, P.; Juul, F.; Srour, B. Ultra-processed foods and cardiometabolic health: Public health policies to reduce consumption cannot wait. BMJ 2023, 383, e075294. [Google Scholar] [CrossRef] [PubMed]
  56. Dicken, S.J.; Batterham, R.L. Ultra-processed food: A global problem requiring a global solution. Lancet Diabetes Endocrinol. 2022, 10, 691–694. [Google Scholar] [CrossRef]
  57. Coletro, H.N.; Bressan, J.; Diniz, A.P.; Hermsdorff, H.H.M.; Pimenta, A.M.; Meireles, A.L.; Mendonca, R.D.; Carraro, J.C.C. Habitual polyphenol intake of foods according to NOVA classification: Implications of ultra-processed foods intake (CUME study). Int. J. Food Sci. Nutr. 2023, 74, 338–349. [Google Scholar] [CrossRef]
  58. Shi, Y.; Yu, C. Effect of dietary living microbe intake on depression symptom in American adult: An opinion from NHANES study. J. Affect. Disord. 2024, 347, 108–114. [Google Scholar] [CrossRef]
  59. Lane, M.M.; Lotfaliany, M.; Hodge, A.M.; O’Neil, A.; Travica, N.; Jacka, F.N.; Rocks, T.; Machado, P.; Forbes, M.; Ashtree, D.N.; et al. High ultra-processed food consumption is associated with elevated psychological distress as an indicator of depression in adults from the Melbourne Collaborative Cohort Study. J. Affect. Disord. 2023, 335, 57–66. [Google Scholar] [CrossRef]
  60. Gomez-Donoso, C.; Sanchez-Villegas, A.; Martinez-Gonzalez, M.A.; Gea, A.; Mendonca, R.D.; Lahortiga-Ramos, F.; Bes-Rastrollo, M. Ultra-processed food consumption and the incidence of depression in a Mediterranean cohort: The SUN Project. Eur. J. Nutr. 2020, 59, 1093–1103. [Google Scholar] [CrossRef]
  61. Adjibade, M.; Julia, C.; Alles, B.; Touvier, M.; Lemogne, C.; Srour, B.; Hercberg, S.; Galan, P.; Assmann, K.E.; Kesse-Guyot, E. Prospective association between ultra-processed food consumption and incident depressive symptoms in the French NutriNet-Sante cohort. BMC Med. 2019, 17, 78. [Google Scholar] [CrossRef]
  62. Lee, S.; Choi, M. Ultra-Processed Food Intakes Are Associated with Depression in the General Population: The Korea National Health and Nutrition Examination Survey. Nutrients 2023, 15, 2169. [Google Scholar] [CrossRef] [PubMed]
  63. Werneck, A.O.; Vancampfort, D.; Oyeyemi, A.L.; Stubbs, B.; Silva, D.R. Joint association of ultra-processed food and sedentary behavior with anxiety-induced sleep disturbance among Brazilian adolescents. J. Affect. Disord. 2020, 266, 135–142. [Google Scholar] [CrossRef] [PubMed]
  64. Coletro, H.N.; Mendonca, R.D.; Meireles, A.L.; Machado-Coelho, G.L.L.; Menezes, M.C. Ultra-processed and fresh food consumption and symptoms of anxiety and depression during the COVID-19 pandemic: COVID Inconfidentes. Clin. Nutr. ESPEN 2022, 47, 206–214. [Google Scholar] [CrossRef] [PubMed]
  65. He, Q.; Wang, Y.; Shen, Y. Association of Ultra-processed Food Consumption with Incident Depression and Anxiety: A Population-based Cohort Study. Food Funct. 2023; in press. [Google Scholar]
  66. Zheng, L.; Sun, J.; Yu, X.; Zhang, D. Ultra-Processed Food Is Positively Associated with Depressive Symptoms Among United States Adults. Front. Nutr. 2020, 7, 600449. [Google Scholar] [CrossRef]
  67. Samuthpongtorn, C.; Nguyen, L.H.; Okereke, O.I.; Wang, D.D.; Song, M.; Chan, A.T.; Mehta, R.S. Consumption of Ultraprocessed Food and Risk of Depression. JAMA Netw. Open 2023, 6, e2334770. [Google Scholar] [CrossRef]
  68. Yeomans, M.R.; Armitage, R.; Atkinson, R.; Francis, H.; Stevenson, R.J. Habitual intake of fat and sugar is associated with poorer memory and greater impulsivity in humans. PLoS ONE 2023, 18, e0290308. [Google Scholar] [CrossRef]
  69. Steele, C.C.; Steele, T.J.; Gwinner, M.; Rosenkranz, S.K.; Kirkpatrick, K. The relationship between dietary fat intake, impulsive choice, and metabolic health. Appetite 2021, 165, 105292. [Google Scholar] [CrossRef]
  70. Epstein, L.H.; Paluch, R.A.; Stein, J.S.; Quattrin, T.; Mastrandrea, L.D.; Bree, K.A.; Sze, Y.Y.; Greenawald, M.H.; Biondolillo, M.J.; Bickel, W.K. Delay discounting, glycemic regulation and health behaviors in adults with prediabetes. Behav. Med. 2021, 47, 194–204. [Google Scholar] [CrossRef]
  71. Zahedi, H.; Kelishadi, R.; Heshmat, R.; Motlagh, M.E.; Ranjbar, S.H.; Ardalan, G.; Payab, M.; Chinian, M.; Asayesh, H.; Larijani, B.; et al. Association between junk food consumption and mental health in a national sample of Iranian children and adolescents: The CASPIAN-IV study. Nutrition 2014, 30, 1391–1397. [Google Scholar] [CrossRef]
  72. Mohseni, H.; Malek Mohammadi, F.; Karampour, Z.; Amini, S.; Abiri, B.; Sayyah, M. The relationship between history of dietary nutrients intakes and incidence of aggressive behavior in adolescent girls: A case-control study. Clin. Nutr. ESPEN 2021, 43, 200–205. [Google Scholar] [CrossRef] [PubMed]
  73. Abiri, B.; Amini, S.; Ehsani, H.; Ehsani, M.; Adineh, P.; Mohammadzadeh, H.; Hashemi, S. Evaluation of dietary food intakes and anthropometric measures in middle-aged men with aggressive symptoms. BMC Nutr. 2023, 9, 75. [Google Scholar] [CrossRef] [PubMed]
  74. Wu, W.C.; Lin, C.I.; Li, Y.F.; Chang, L.Y.; Chiang, T.L. The mediating effect of dietary patterns on the association between mother’s education level and the physical aggression of five-year-old children: A population-based cohort study. BMC Pediatr. 2020, 20, 221. [Google Scholar] [CrossRef] [PubMed]
  75. Khayyatzadeh, S.S.; Firouzi, S.; Askari, M.; Mohammadi, F.; Nikbakht-Jam, I.; Ghazimoradi, M.; Mohammadzadeh, M.; Ferns, G.A.; Ghayour-Mobarhan, M. Dietary intake of carotenoids and fiber is inversely associated with aggression score in adolescent girls. Nutr. Health 2019, 25, 203–208. [Google Scholar] [CrossRef]
  76. Mrug, S.; Jones, L.C.; Elliott, M.N.; Tortolero, S.R.; Peskin, M.F.; Schuster, M.A. Soft Drink Consumption and Mental Health in Adolescents: A Longitudinal Examination. J. Adolesc. Health 2021, 68, 155–160. [Google Scholar] [CrossRef]
  77. Gketsios, I.; Tsiampalis, T.; Kanellopoulou, A.; Vassilakou, T.; Notara, V.; Antonogeorgos, G.; Rojas-Gil, A.P.; Kornilaki, E.N.; Lagiou, A.; Panagiotakos, D.B.; et al. The Synergetic Effect of Soft Drinks and Sweet/Salty Snacks Consumption and the Moderating Role of Obesity on Preadolescents’ Emotions and Behavior: A School-Based Epidemiological Study. Life 2023, 13, 633. [Google Scholar] [CrossRef]
  78. Drouka, A.; Mamalaki, E.; Karavasilis, E.; Scarmeas, N.; Yannakoulia, M. Dietary and nutrient patterns and brain MRI biomarkers in dementia-free adults. Nutrients 2022, 14, 2345. [Google Scholar] [CrossRef]
  79. Akbaraly, T.; Sexton, C.; Zsoldos, E.; Mahmood, A.; Filippini, N.; Kerleau, C.; Verdier, J.-M.; Virtanen, M.; Gabelle, A.; Ebmeier, K.P. Association of long-term diet quality with hippocampal volume: Longitudinal cohort study. Am. J. Med. 2018, 131, 1372–1381.e4. [Google Scholar] [CrossRef] [PubMed]
  80. Attuquayefio, T.; Stevenson, R.J.; Oaten, M.J.; Francis, H.M. A four-day Western-style dietary intervention causes reductions in hippocampal-dependent learning and memory and interoceptive sensitivity. PLoS ONE 2017, 12, e0172645. [Google Scholar] [CrossRef]
  81. Hall, K.D.; Ayuketah, A.; Brychta, R.; Cai, H.; Cassimatis, T.; Chen, K.Y.; Chung, S.T.; Costa, E.; Courville, A.; Darcey, V.; et al. Ultra-Processed Diets Cause Excess Calorie Intake and Weight Gain: An Inpatient Randomized Controlled Trial of Ad Libitum Food Intake. Cell. Metab. 2019, 30, 226. [Google Scholar] [CrossRef]
  82. O’Connor, L.E.; Hall, K.D.; Herrick, K.A.; Reedy, J.; Chung, S.T.; Stagliano, M.; Courville, A.B.; Sinha, R.; Freedman, N.D.; Hong, H.G.; et al. Metabolomic Profiling of an Ultraprocessed Dietary Pattern in a Domiciled Randomized Controlled Crossover Feeding Trial. J. Nutr. 2023, 153, 2181–2192. [Google Scholar] [CrossRef] [PubMed]
  83. Gearhardt, A.N.; Bueno, N.B.; DiFeliceantonio, A.G.; Roberto, C.A.; Jimenez-Murcia, S.; Fernandez-Aranda, F. Social, clinical, and policy implications of ultra-processed food addiction. BMJ 2023, 383, e075354. [Google Scholar] [CrossRef] [PubMed]
  84. Delgado-Rodríguez, R.; Moreno-Padilla, M.; Moreno-Domínguez, S.; Cepeda-Benito, A. Food Addiction Correlates with Emotional and Craving Reactivity to Industrially Prepared (Ultra-Processed) and Home-Cooked (Processed) Foods but not Unprocessed or Minimally Processed Foods. Food Qual. Prefer. 2023, 110, 104961. [Google Scholar] [CrossRef]
  85. Tristan Asensi, M.; Napoletano, A.; Sofi, F.; Dinu, M. Low-Grade Inflammation and Ultra-Processed Foods Consumption: A Review. Nutrients 2023, 15, 1546. [Google Scholar] [CrossRef]
  86. Lane, M.M.; Lotfaliany, M.; Forbes, M.; Loughman, A.; Rocks, T.; O’Neil, A.; Machado, P.; Jacka, F.N.; Hodge, A.; Marx, W. Higher Ultra-Processed Food Consumption Is Associated with Greater High-Sensitivity C-Reactive Protein Concentration in Adults: Cross-Sectional Results from the Melbourne Collaborative Cohort Study. Nutrients 2022, 14, 3309. [Google Scholar] [CrossRef]
  87. Li, H.; Wang, Y.; Sonestedt, E.; Borne, Y. Associations of ultra-processed food consumption, circulating protein biomarkers, and risk of cardiovascular disease. BMC Med. 2023, 21, 415. [Google Scholar] [CrossRef]
  88. Corbin, K.D.; Carnero, E.A.; Dirks, B.; Igudesman, D.; Yi, F.; Marcus, A.; Davis, T.L.; Pratley, R.E.; Rittmann, B.E.; Krajmalnik-Brown, R.; et al. Host-diet-gut microbiome interactions influence human energy balance: A randomized clinical trial. Nat. Commun. 2023, 14, 3161. [Google Scholar] [CrossRef]
  89. Zhang, P. Influence of Foods and Nutrition on the Gut Microbiome and Implications for Intestinal Health. Int. J. Mol. Sci. 2022, 23, 9588. [Google Scholar] [CrossRef]
  90. Yu, G.; Xu, C.; Zhang, D.; Ju, F.; Ni, Y. MetOrigin: Discriminating the origins of microbial metabolites for integrative analysis of the gut microbiome and metabolome. IMeta 2022, 1, e10. [Google Scholar] [CrossRef]
  91. Atzeni, A.; Martinez, M.A.; Babio, N.; Konstanti, P.; Tinahones, F.J.; Vioque, J.; Corella, D.; Fito, M.; Vidal, J.; Moreno-Indias, I.; et al. Association between ultra-processed food consumption and gut microbiota in senior subjects with overweight/obesity and metabolic syndrome. Front. Nutr. 2022, 9, 976547. [Google Scholar] [CrossRef]
  92. Khiroya, K.; Sekyere, E.; McEwen, B.; Bayes, J. Nutritional considerations in major depressive disorder: Current evidence and functional testing for clinical practice. Nutr. Res. Rev. 2023, 1–12. [Google Scholar] [CrossRef] [PubMed]
  93. Winiarska-Mieczan, A.; Kwiecien, M.; Jachimowicz-Rogowska, K.; Donaldson, J.; Tomaszewska, E.; Baranowska-Wojcik, E. Anti-Inflammatory, Antioxidant, and Neuroprotective Effects of Polyphenols-Polyphenols as an Element of Diet Therapy in Depressive Disorders. Int. J. Mol. Sci. 2023, 24, 2258. [Google Scholar] [CrossRef] [PubMed]
  94. Marx, W.; Lane, M.; Hockey, M.; Aslam, H.; Berk, M.; Walder, K.; Borsini, A.; Firth, J.; Pariante, C.M.; Berding, K.; et al. Diet and depression: Exploring the biological mechanisms of action. Mol. Psychiatry 2021, 26, 134–150. [Google Scholar] [CrossRef]
  95. Ortega, M.; Fraile-Martínez, Ó.; García-Montero, C.; Alvarez-Mon, M.; Lahera, G.; Monserrat, J.; Llavero-Valero, M.; Gutiérrez-Rojas, L.; Molina, R.; Rodríguez-Jimenez, R. Biological role of nutrients, food and dietary patterns in the prevention and clinical management of major depressive disorder. Nutrients 2022, 14, 3099. [Google Scholar] [CrossRef] [PubMed]
  96. Contreras-Rodriguez, O.; Solanas, M.; Escorihuela, R.M. Dissecting ultra-processed foods and drinks: Do they have a potential to impact the brain? Rev. Endocr. Metab. Disord. 2022, 23, 697–717. [Google Scholar] [CrossRef]
  97. Bhave, V.M.; Oladele, C.R.; Ament, Z.; Kijpaisalratana, N.; Jones, A.C.; Couch, C.A.; Patki, A.; Garcia Guarniz, A.-L.; Bennett, A.; Crowe, M. Associations Between Ultra-Processed Food Consumption and Adverse Brain Health Outcomes. Neurology 2024, 102, e209432. [Google Scholar] [CrossRef] [PubMed]
  98. Contreras-Rodriguez, O.; Reales-Moreno, M.; Fernandez-Barres, S.; Cimpean, A.; Arnoriaga-Rodriguez, M.; Puig, J.; Biarnes, C.; Motger-Alberti, A.; Cano, M.; Fernandez-Real, J.M. Consumption of ultra-processed foods is associated with depression, mesocorticolimbic volume, and inflammation. J. Affect. Disord. 2023, 335, 340–348. [Google Scholar] [CrossRef]
  99. Ued, F.V.; Mathias, M.G.; Toffano, R.B.D.; Barros, T.T.; Almada, M.; Salomao, R.G.; Coelho-Landell, C.A.; Hillesheim, E.; Camarneiro, J.M.; Camelo-Junior, J.S.; et al. Vitamin B2 and Folate Concentrations are Associated with ARA, EPA and DHA Fatty Acids in Red Blood Cells of Brazilian Children and Adolescents. Nutrients 2019, 11, 2918. [Google Scholar] [CrossRef]
  100. Portnoy, J.; McGouldrick, S.H.; Raine, A.; Zemel, B.S.; Tucker, K.L.; Liu, J. Lower dietary intake of magnesium is associated with more callous-unemotional traits in children. Nutr. Neurosci. 2022, 25, 2314–2323. [Google Scholar] [CrossRef]
  101. Fiorentini, D.; Cappadone, C.; Farruggia, G.; Prata, C. Magnesium: Biochemistry, nutrition, detection, and social impact of diseases linked to its deficiency. Nutrients 2021, 13, 1136. [Google Scholar] [CrossRef]
  102. Gurgen, S.G.; Sayın, O.; Çetin, F.; Sarsmaz, H.Y.; Yazıcı, G.l.N.; Umur, N.; Yucel, A.T. The effect of monosodium glutamate on neuronal signaling molecules in the hippocampus and the neuroprotective effects of omega-3 fatty acids. ACS Chemical. Neurosci. 2021, 12, 3028–3037. [Google Scholar] [CrossRef] [PubMed]
  103. Lopresti, A.L. The effects of psychological and environmental stress on micronutrient concentrations in the body: A review of the evidence. Adv. Nutr. 2020, 11, 103–112. [Google Scholar] [CrossRef]
  104. Coccaro, E.F.; Lee, R.; Breen, E.C.; Irwin, M.R. Plasma and cerebrospinal fluid inflammatory markers and human aggression. Neuropsychopharmacol 2023, 48, 1060–1066. [Google Scholar] [CrossRef]
  105. Blomström, Å.; Kosidou, K.; Kristiansson, M.; Masterman, T. Infection during childhood and the risk of violent criminal behavior in adulthood. Brain Behav. Immun. 2020, 86, 63–71. [Google Scholar] [CrossRef] [PubMed]
  106. Takahashi, A.; Flanigan, M.E.; McEwen, B.S.; Russo, S.J. Aggression, social stress, and the immune system in humans and animal models. Front. Behav. Neurosci. 2018, 12, 338283. [Google Scholar] [CrossRef]
  107. Flux, M.; Lowry, C.A. Inflammation as a mediator of stress-related psychiatric disorders. In Neurobiology of Brain Disorders: Biological Basis of Neurological and Psychiatric Disorders; Zigmond, M.J., Wiley, C.A., Chesselet, M.-F., Eds.; Academic Press: Cambridge, MA, USA, 2023; pp. 885–911. [Google Scholar]
  108. Takahashi, A. Associations of the immune system in aggression traits and the role of microglia as mediators. Neuropharmacology 2024, 256, 110021. [Google Scholar] [CrossRef]
  109. Luo, Y. The crosstalk between the “inflamed” mind and the “impulsive” mind: Activation of microglia and impulse control disorders. In Proceedings of the Second International Conference on Biological Engineering and Medical Science (ICBioMed 2022), Oxford, UK, 7–13 November 2022; pp. 650–665. [Google Scholar]
  110. Li, H.; Watkins, L.R.; Wang, X. Microglia in neuroimmunopharmacology and drug addiction. Mol. Psychiatry 2024, 29, 1912–1924. [Google Scholar] [CrossRef] [PubMed]
  111. Holton, K.F. Micronutrients may be a unique weapon against the neurotoxic triad of excitotoxicity, oxidative stress and neuroinflammation: A perspective. Front. Neurosci. 2021, 15, 726457. [Google Scholar] [CrossRef]
  112. Erickson, M.A.; Dohi, K.; Banks, W.A. Neuroinflammation: A common pathway in CNS diseases as mediated at the blood-brain barrier. Neuroimmunomodulation 2012, 19, 121–130. [Google Scholar] [CrossRef]
  113. Holton, K. The potential role of dietary intervention for the treatment of neuroinflammation. In Translational Neuroimmunology, Volume 7; Academic Press: Cambridge, MA, USA, 2023; pp. 239–266. [Google Scholar]
  114. Nukina, H.; Sudo, N.; Aiba, Y.; Oyama, N.; Koga, Y.; Kubo, C. Restraint stress elevates the plasma interleukin-6 levels in germ-free mice. J. Neuroimmunol. 2001, 115, 46–52. [Google Scholar] [CrossRef]
  115. Bostick, J.W.; Schonhoff, A.M.; Mazmanian, S.K. Gut microbiome-mediated regulation of neuroinflammation. Curr. Opin. Immunol. 2022, 76, 102177. [Google Scholar] [CrossRef] [PubMed]
  116. Zhao, Q.; Baranova, A.; Cao, H.; Zhang, F. Gut microbiome and major depressive disorder: Insights from two-sample Mendelian randomization. BMC Psychiatry 2024, 24, 493. [Google Scholar] [CrossRef] [PubMed]
  117. Faulkner, P.; Costabile, A.; Imakulata, F.; Pandey, N.; Hepsomali, P. A preliminary examination of gut microbiota and emotion regulation in 2-to 6-year-old children. Front. Dev. Psychol. 2024, 2, 1445642. [Google Scholar] [CrossRef]
  118. Devason, A.S.; Thaiss, C.A.; de la Fuente-Nunez, C. Neuromicrobiology Comes of Age: The Multifaceted Interactions between the Microbiome and the Nervous System. ACS Chem. Neurosci. 2024, 15, 2957–2965. [Google Scholar] [CrossRef]
  119. Fernandes, A.E.; Rosa, P.W.; Melo, M.E.; Martins, R.C.; Santin, F.G.; Moura, A.M.; Coelho, G.S.; Sabino, E.C.; Cercato, C.; Mancini, M.C. Differences in the gut microbiota of women according to ultra-processed food consumption. Nutr. Metab. Cardiovasc. Dis. 2023, 33, 84–89. [Google Scholar] [CrossRef]
  120. Song, Z.; Song, R.; Liu, Y.; Wu, Z.; Zhang, X. Effects of ultra-processed foods on the microbiota-gut-brain axis: The bread-and-butter issue. Food Res. Int. 2023, 167, 112730. [Google Scholar] [CrossRef]
  121. Martinez Leo, E.E.; Segura Campos, M.R. Effect of ultra-processed diet on gut microbiota and thus its role in neurodegenerative diseases. Nutrition 2020, 71, 110609. [Google Scholar] [CrossRef] [PubMed]
  122. Naimi, S.; Viennois, E.; Gewirtz, A.T.; Chassaing, B. Direct impact of commonly used dietary emulsifiers on human gut microbiota. Microbiome 2021, 9, 66. [Google Scholar] [CrossRef]
  123. Kyaw, T.S.; Sukmak, M.; Nahok, K.; Sharma, A.; Silsirivanit, A.; Lert-Itthiporn, W.; Sansurin, N.; Senthong, V.; Anutrakulchai, S.; Sangkhamanon, S.; et al. Monosodium glutamate consumption reduces the renal excretion of trimethylamine N-oxide and the abundance of Akkermansia muciniphila in the gut. Biochem. Biophys. Res. Commun. 2022, 630, 158–166. [Google Scholar] [CrossRef]
  124. Berding, K.; Bastiaanssen, T.F.S.; Moloney, G.M.; Boscaini, S.; Strain, C.R.; Anesi, A.; Long-Smith, C.; Mattivi, F.; Stanton, C.; Clarke, G.; et al. Feed your microbes to deal with stress: A psychobiotic diet impacts microbial stability and perceived stress in a healthy adult population. Mol. Psychiatry 2023, 28, 601–610. [Google Scholar] [CrossRef]
  125. Magzal, F.; Turroni, S.; Fabbrini, M.; Barone, M.; Vitman Schorr, A.; Ofran, A.; Tamir, S. A personalized diet intervention improves depression symptoms and changes microbiota and metabolite profiles among community-dwelling older adults. Front. Nutr. 2023, 10, 1234549. [Google Scholar] [CrossRef] [PubMed]
  126. Bruce-Keller, A.J.; Salbaum, J.M.; Luo, M.; Blanchard, E.T.; Taylor, C.M.; Welsh, D.A.; Berthoud, H.R. Obese-type gut microbiota induce neurobehavioral changes in the absence of obesity. Biol. Psychiatry 2015, 77, 607–615. [Google Scholar] [CrossRef] [PubMed]
  127. Yang, Y.; Zhong, Z.; Wang, B.; Xia, X.; Yao, W.; Huang, L.; Wang, Y.; Ding, W. Early-life high-fat diet-induced obesity programs hippocampal development and cognitive functions via regulation of gut commensal Akkermansia muciniphila. Neuropsychopharmacol 2019, 44, 2054–2064. [Google Scholar] [CrossRef] [PubMed]
  128. Duan, C.; Huang, L.; Zhang, C.; Zhang, L.; Xia, X.; Zhong, Z.; Wang, B.; Wang, Y.; Man Hoi, M.P.; Ding, W.; et al. Gut commensal-derived butyrate reverses obesity-induced social deficits and anxiety-like behaviors via regulation of microglial homeostasis. Eur. J. Pharmacol. 2021, 908, 174338. [Google Scholar] [CrossRef]
  129. Wei, N.; Ju, M.; Su, X.; Zhang, Y.; Huang, Y.; Rao, X.; Cui, L.; Lin, Z.; Dong, Y. Transplantation of gut microbiota derived from patients with schizophrenia induces schizophrenia-like behaviors and dysregulated brain transcript response in mice. Schizophrenia 2024, 10, 44. [Google Scholar] [CrossRef] [PubMed]
  130. Xiao, L.; Yan, J.; Yang, T.; Zhu, J.; Li, T.; Wei, H.; Chen, J. Fecal Microbiome Transplantation from Children with Autism Spectrum Disorder Modulates Tryptophan and Serotonergic Synapse Metabolism and Induces Altered Behaviors in Germ-Free Mice. mSystems 2021, 6, e01343-20. [Google Scholar] [CrossRef]
  131. Kelly, J.R.; Borre, Y.; O’Brien, C.; Patterson, E.; El Aidy, S.; Deane, J.; Kennedy, P.J.; Beers, S.; Scott, K.; Moloney, G.; et al. Transferring the blues: Depression-associated gut microbiota induces neurobehavioural changes in the rat. J. Psychiatr. Res. 2016, 82, 109–118. [Google Scholar] [CrossRef]
  132. Wolstenholme, J.T.; Saunders, J.M.; Smith, M.; Kang, J.D.; Hylemon, P.B.; Gonzalez-Maeso, J.; Fagan, A.; Zhao, D.; Sikaroodi, M.; Herzog, J.; et al. Reduced alcohol preference and intake after fecal transplant in patients with alcohol use disorder is transmissible to germ-free mice. Nat. Commun. 2022, 13, 6198. [Google Scholar] [CrossRef]
  133. Wang, T.; Hao, L.; Yang, K.; Feng, W.; Guo, Z.; Liu, M.; Xiao, R. Fecal microbiota transplantation derived from mild cognitive impairment individuals impairs cerebral glucose uptake and cognitive function in wild-type mice: Bacteroidetes and TXNIP-GLUT signaling pathway. Gut. Microbes. 2024, 16, 2395907. [Google Scholar] [CrossRef]
  134. Wang, C.; Yan, J.; Du, K.; Liu, S.; Wang, J.; Wang, Q.; Zhao, H.; Li, M.; Yan, D.; Zhang, R.; et al. Intestinal microbiome dysbiosis in alcohol-dependent patients and its effect on rat behaviors. mBio 2023, 14, e0239223. [Google Scholar] [CrossRef]
  135. Uzan-Yulzari, A.; Turjeman, S.; Moadi, L.; Getselter, D.; Rautava, S.; Isolauri, E.; Khatib, S.; Elliott, E.; Koren, O. A gut reaction? The role of the microbiome in aggression. Brain Behav. Immun. 2024, 122, 301–312. [Google Scholar] [CrossRef] [PubMed]
  136. Zhang, H.; Wei, H.; Qin, X.; Song, H.; Yang, M.; Zhang, L.; Liu, Y.; Wang, Z.; Zhang, Y.; Lai, Y. Is anxiety and depression transmissible?—Depressed mother rats transmit anxiety-and depression-like phenotypes to cohabited rat pups through gut microbiota assimilation. J. Affect. Disord. 2024, 366, 124–135. [Google Scholar] [CrossRef] [PubMed]
  137. Vanuytsel, T.; van Wanrooy, S.; Vanheel, H.; Vanormelingen, C.; Verschueren, S.; Houben, E.; Salim Rasoel, S.; Tomicronth, J.; Holvoet, L.; Farre, R.; et al. Psychological stress and corticotropin-releasing hormone increase intestinal permeability in humans by a mast cell-dependent mechanism. Gut 2014, 63, 1293–1299. [Google Scholar] [CrossRef] [PubMed]
  138. Li, X.; Kan, E.M.; Lu, J.; Cao, Y.; Wong, R.K.; Keshavarzian, A.; Wilder-Smith, C.H. Combat-training increases intestinal permeability, immune activation and gastrointestinal symptoms in soldiers. Aliment. Pharm. Ther. 2013, 37, 799–809. [Google Scholar] [CrossRef] [PubMed]
  139. Flux, M.; Lowry, C.A. Finding intestinal fortitude: Integrating the microbiome into a holistic view of depression mechanisms, treatment, and resilience. Neurobiol. Dis. 2020, 135, 104578. [Google Scholar] [CrossRef] [PubMed]
  140. Noble, E.E.; Hsu, T.M.; Kanoski, S.E. Gut to Brain Dysbiosis: Mechanisms Linking Western Diet Consumption, the Microbiome, and Cognitive Impairment. Front. Behav. Neurosci. 2017, 11, 9. [Google Scholar] [CrossRef]
  141. Brouillet, J.Z.; Boltri, M.; Lengvenyte, A.; Lajnef, M.; Richard, J.R.; Barrau, C.; Tamouza, R. Association of markers of inflammation and intestinal permeability in suicidal patients with major mood disorders. J. Affect. Disord. Rep. 2023, 14, 100624. [Google Scholar] [CrossRef]
  142. Voigt, R.M.; Zalta, A.K.; Raeisi, S.; Zhang, L.; Brown, J.M.; Forsyth, C.B.; Boley, R.A.; Held, P.; Pollack, M.H.; Keshavarzian, A. Abnormal intestinal milieu in posttraumatic stress disorder is not impacted by treatment that improves symptoms. Am. J. Physiol. Gastrointest. Liver Physiol. 2022, 323, G61–G70. [Google Scholar] [CrossRef]
  143. Asejeje, F.O.; Abiola, M.A.; Adeyemo, O.A.; Ogunro, O.B.; Ajayi, A.M. Exogenous monosodium glutamate exacerbates lipopolysaccharide-induced neurobehavioral deficits, oxidative damage, neuroinflammation, and cholinergic dysfunction in rat brain. Neurosci. Lett. 2024, 825, 137710. [Google Scholar] [CrossRef]
  144. Abdelhamid, W.G.; Mowaad, N.A.; Asaad, G.F.; Galal, A.F.; Mohammed, S.S.; Mostafa, O.E.; Sadek, D.R.; Elkhateb, L.A. The potential protective effect of Camellia Sinensis in mitigating monosodium glutamate-induced neurotoxicity: Biochemical and histological study in male albino rats. Metab. Brain Dis. 2024, 39, 953–966. [Google Scholar] [CrossRef]
  145. Abu-Elfotuh, K.; Abdel-Sattar, S.A.; Abbas, A.N.; Mahran, Y.F.; Alshanwani, A.R.; Hamdan, A.M.E.; Atwa, A.M.; Reda, E.; Ahmed, Y.M.; Zaghlool, S.S. The protective effect of thymoquinone or/and thymol against monosodium glutamate-induced attention-deficit/hyperactivity disorder (ADHD)-like behavior in rats: Modulation of Nrf2/HO-1, TLR4/NF-κB/NLRP3/caspase-1 and Wnt/β-Catenin signaling pathways in rat model. Biomed. Pharmacother. 2022, 155, 113799. [Google Scholar]
  146. Ahangari, H.; Bahramian, B.; Khezerlou, A.; Tavassoli, M.; Kiani-Salmi, N.; Tarhriz, V.; Ehsani, A. Association between monosodium glutamate consumption with changes in gut microbiota and related metabolic dysbiosis—A systematic review. Food Sci. Nutr. 2024, 12, 5285–5295. [Google Scholar] [CrossRef] [PubMed]
  147. Gasperotti, M.; Passamonti, S.; Tramer, F.; Masuero, D.; Guella, G.; Mattivi, F.; Vrhovsek, U. Fate of microbial metabolites of dietary polyphenols in rats: Is the brain their target destination? ACS Chem. Neurosci. 2015, 6, 1341–1352. [Google Scholar] [CrossRef] [PubMed]
  148. Xue, F.; He, Z.; Zhuang, D.Z.; Lin, F. The influence of gut microbiota on circulating inflammatory cytokines and host: A Mendelian randomization study with meta-analysis. Life Sci. 2023, 332, 122105. [Google Scholar] [CrossRef]
  149. Wang, C.; Zhang, T.; He, L.; Fu, J.Y.; Deng, H.X.; Xue, X.L.; Chen, B.T. Bacterial Translocation Associates With Aggression in Schizophrenia Inpatients. Front. Syst. Neurosci. 2021, 15, 704069. [Google Scholar] [CrossRef]
  150. Deng, H.; He, L.; Wang, C.; Zhang, T.; Guo, H.; Zhang, H.; Song, Y.; Chen, B. Altered gut microbiota and its metabolites correlate with plasma cytokines in schizophrenia inpatients with aggression. BMC Psychiatry 2022, 22, 629. [Google Scholar] [CrossRef]
  151. Russo, S.; Chan, K.; Li, L.; Parise, L.; Cathomas, F.; LeClair, K.; Shimo, Y.; Lin, H.Y.; Durand-de Cuttoli, R.; Aubry, A.; et al. Stress-activated brain-gut circuits disrupt intestinal barrier integrity and social behaviour. Res. Sq. 2023. [Google Scholar] [CrossRef]
  152. Zeamer, A.L.; Salive, M.C.; An, X.; Beaudoin, F.L.; House, S.L.; Stevens, J.S.; Zeng, D.; Neylan, T.C.; Clifford, G.D.; Linnstaedt, S.D.; et al. Association between microbiome and the development of adverse posttraumatic neuropsychiatric sequelae after traumatic stress exposure. Transl. Psychiatry 2023, 13, 354. [Google Scholar] [CrossRef]
  153. Jakobi, B.; Cimetti, C.; Mulder, D.; Vlaming, P.; Franke, B.; Hoogman, M.; Arias-Vasquez, A. The Role of Diet and the Gut Microbiota in Reactive Aggression and Adult ADHD-An Exploratory Analysis. Nutrients 2024, 16, 2174. [Google Scholar] [CrossRef]
  154. Meijnikman, A.S.; Nieuwdorp, M.; Schnabl, B. Endogenous ethanol production in health and disease. Nat. Rev. Gastroenterol. Hepatol. 2024, 21, 556–571. [Google Scholar] [CrossRef]
  155. Paton, A. Alcohol in the body. BMJ 2005, 330, 85–87. [Google Scholar] [CrossRef] [PubMed]
  156. Robinson, J.M.; Wissel, E.F.; Breed, M.F. Policy implications of the microbiota-gut-brain axis. Trends Microbiol. 2024, 32, 107–110. [Google Scholar] [CrossRef] [PubMed]
  157. Schoenthaler, S.J. The effect of sugar on the treatment and control of antisocial behavior: A double-blind study of an incarcerated juvenile population. Int. J. Biosoc. Res. 1982, 3, 1–9. [Google Scholar]
  158. Schoenthaler, S.J. Diet and crime: An empirical examination of the value of nutrition in the control and treatment of incarcerated juvenile offenders. Int. J. Biosoc. Res. 1983, 4, 25–39. [Google Scholar] [CrossRef]
  159. Schoenthaler, S.J. The Los Angeles Probation Department diet-behavior program: An empirical analysis of six institutional settings. Int. J. Biosoc. Res. 1983, 5, 88–98. [Google Scholar] [CrossRef]
  160. Schoenthaler, S.J. The Alabama diet-behavior program: An empirical evaluation at the Coosa Valley Regional Detention Center. Int. J. Biosoc. Res. 1983, 5, 79–87. [Google Scholar] [CrossRef]
  161. Schoenthaler, S.J. Diet Crime and Delinquency-A Review of the 1983 and 1984 Studies. Int. J. Biosoc. Res. 1984, 6, 141–153. [Google Scholar]
  162. Schoenthaler, S.J.; Bier, I.D. Diet and delinquency: Empirical testing of seven theories. Int. J. Biosoc. Res. 1985, 7, 108–131. [Google Scholar] [CrossRef]
  163. Giannouli, V.; Stamovlasis, D.; Tsolaki, M. Longitudinal study of depression on amnestic mild cognitive impairment and financial capacity. Clin. Gerontol. 2022, 45, 708–714. [Google Scholar] [CrossRef]
  164. Giannouli, V.; Tsolaki, M. Vascular dementia, depression, and financial capacity assessment. Alzheimer Dis. Assoc. Disord. 2021, 35, 84–87. [Google Scholar] [CrossRef]
  165. Giannouli, V.; Tsolaki, M. Unraveling Ariadne’s thread into the labyrinth of aMCI: Depression and financial capacity. Alzheimer Dis. Assoc. Disord. 2021, 35, 363–365. [Google Scholar] [CrossRef] [PubMed]
  166. Giannouli, V.; Tsolaki, M. Mild Alzheimer Disease, financial capacity, and the role of depression: Eyes wide shut? Alzheimer Dis. Assoc. Disord. 2021, 35, 360–362. [Google Scholar] [CrossRef]
  167. Giannouli, V.; Tsolaki, M. Is depression or apathy playing a key role in predicting financial capacity in Parkinson’s Disease with Dementia and Frontotemporal Dementia? Brain Sci. 2021, 11, 785. [Google Scholar] [CrossRef] [PubMed]
  168. Giannouli, V.; Tsolaki, M. Beneath the Top of the Iceberg: Financial Capacity Deficits in Mixed Dementia with and without Depression. Healthcare 2023, 11, 505. [Google Scholar] [CrossRef]
  169. Lu, J.; Zhao, X.; Wei, X.; He, G. Risky decision-making in major depressive disorder: A three-level meta-analysis. Int. J. Clin. Health Psychol. 2024, 24, 100417. [Google Scholar] [CrossRef]
  170. Noreen, S.; Dritschel, B. In the here and now: Future thinking and social problem-solving in depression. PLoS ONE 2022, 17, e0270661. [Google Scholar] [CrossRef] [PubMed]
  171. Jacka, F.N.; O’Neil, A.; Opie, R.; Itsiopoulos, C.; Cotton, S.; Mohebbi, M.; Castle, D.; Dash, S.; Mihalopoulos, C.; Chatterton, M.L.; et al. A randomised controlled trial of dietary improvement for adults with major depression (the ‘SMILES’ trial). BMC Med. 2017, 15, 23. [Google Scholar] [CrossRef] [PubMed]
  172. Lane, M.; Lotfaliany, M.; Machado, P.; Jacka, F.; Mohebbi, M.; O’Neil, A.; Werneck, A.O.; Monteiro, C.; Loughman, A.; Rocks, T.; et al. Change in Ultra-Processed Food Consumption Moderates Clinical Trial Outcomes in Depression: A Secondary Analysis of the SMILES Randomised Controlled Trial. Preprints 2023, 2023081110. [Google Scholar] [CrossRef]
  173. Francis, H.M.; Stevenson, R.J.; Chambers, J.R.; Gupta, D.; Newey, B.; Lim, C.K. A brief diet intervention can reduce symptoms of depression in young adults—A randomised controlled trial. PLoS ONE 2019, 14, e0222768. [Google Scholar] [CrossRef]
  174. Agarwal, U.; Mishra, S.; Xu, J.; Levin, S.; Gonzales, J.; Barnard, N.D. A multicenter randomized controlled trial of a nutrition intervention program in a multiethnic adult population in the corporate setting reduces depression and anxiety and improves quality of life: The GEICO study. Am. J. Health. Promot. 2015, 29, 245–254. [Google Scholar] [CrossRef]
  175. Parletta, N.; Zarnowiecki, D.; Cho, J.; Wilson, A.; Bogomolova, S.; Villani, A.; Itsiopoulos, C.; Niyonsenga, T.; Blunden, S.; Meyer, B.; et al. A Mediterranean-style dietary intervention supplemented with fish oil improves diet quality and mental health in people with depression: A randomized controlled trial (HELFIMED). Nutr. Neurosci. 2019, 22, 474–487. [Google Scholar] [CrossRef] [PubMed]
  176. Bayes, J.; Schloss, J.; Sibbritt, D. The effect of a Mediterranean diet on the symptoms of depression in young males (the “AMMEND: A Mediterranean Diet in MEN with Depression” study): A randomized controlled trial. Am. J. Clin. Nutr. 2022, 116, 572–580. [Google Scholar] [CrossRef] [PubMed]
  177. Cabrera-Suarez, B.M.; Lahortiga-Ramos, F.; Sayon-Orea, C.; Hernandez-Fleta, J.L.; Gonzalez-Pinto, A.; Molero, P.; Vega-Perez, R.; Sanchez-Villegas, A.; Cabrera, C.; Pla, J.; et al. Effect of a dietary intervention based on the Mediterranean diet on the quality of life of patients recovered from depression: Analysis of the PREDIDEP randomized trial. Exp. Gerontol. 2023, 175, 112149. [Google Scholar] [CrossRef] [PubMed]
  178. McMillan, L.; Owen, L.; Kras, M.; Scholey, A. Behavioural effects of a 10-day Mediterranean diet. Results from a pilot study evaluating mood and cognitive performance. Appetite 2011, 56, 143–147. [Google Scholar] [CrossRef] [PubMed]
  179. Firth, J.; Marx, W.; Dash, S.; Carney, R.; Teasdale, S.B.; Solmi, M.; Stubbs, B.; Schuch, F.B.; Carvalho, A.F.; Jacka, F. The effects of dietary improvement on symptoms of depression and anxiety: A meta-analysis of randomized controlled trials. Psychosom. Med. 2019, 81, 265–280. [Google Scholar] [CrossRef]
  180. Schoenthaler, S.J. The effects of citrus on the treatment and control of antisocial behavior: A double-blind study of an incarcerated juvenile population. Int. J. Biosoc. Res. 1983, 5, 107–117. [Google Scholar] [CrossRef]
  181. Choi, J.; Kim, J.H.; Park, M.; Lee, H.J. Effects of Flavonoid-Rich Orange Juice Intervention on Major Depressive Disorder in Young Adults: A Randomized Controlled Trial. Nutrients 2022, 15, 145. [Google Scholar] [CrossRef]
  182. Gillies, N.A.; Wilson, B.C.; Miller, J.R.; Roy, N.C.; Scholey, A.; Braakhuis, A.J. Effects of a Flavonoid-Rich Blackcurrant Beverage on Markers of the Gut-Brain Axis in Healthy Females: Secondary Findings From a 4-Week Randomized Crossover Control Trial. Curr. Dev. Nutr. 2024, 8, 102158. [Google Scholar] [CrossRef]
  183. Maury, A.A.; Holton, K.F. Biomarkers Associated with Depression Improvement in Veterans with Gulf War Illness Using the Low-Glutamate Diet. Nutrients 2024, 16, 2255. [Google Scholar] [CrossRef]
  184. Murray, S.L.; Holton, K.F. Effects of a diet low in excitotoxins on PTSD symptoms and related biomarkers. Nutr. Neurosci. 2022, 27, 1–11. [Google Scholar] [CrossRef]
  185. Shinn, L.M.; Mansharamani, A.; Baer, D.J.; Novotny, J.A.; Charron, C.S.; Khan, N.A.; Zhu, R.; Holscher, H.D. Fecal Metagenomics to Identify Biomarkers of Food Intake in Healthy Adults: Findings from Randomized, Controlled, Nutrition Trials. J. Nutr. 2024, 154, 271–283. [Google Scholar] [CrossRef]
  186. Raine, A.; Brodrick, L. Omega-3 supplementation reduces aggressive behavior: A meta-analytic review of randomized controlled trials. Aggress. Violent Behav. 2024, 78, 101956. [Google Scholar] [CrossRef] [PubMed]
  187. Gesch, C.B.; Hammond, S.M.; Hampson, S.E.; Eves, A.; Crowder, M.J. Influence of supplementary vitamins, minerals and essential fatty acids on the antisocial behaviour of young adult prisoners: Randomised, placebo-controlled trial. Br. J. Psychiatry 2002, 181, 22–28. [Google Scholar] [CrossRef] [PubMed]
  188. Choy, O. Nutritional factors associated with aggression. Front. Psychiatry 2023, 14, 1176061. [Google Scholar] [CrossRef] [PubMed]
  189. Lai, W.D.; Tung, T.H.; Teng, C.Y.; Chang, C.H.; Chen, Y.C.; Huang, H.Y.; Lee, H.C.; Huang, S.Y. Fish oil ameliorates neuropsychiatric behaviors and gut dysbiosis by elevating selected microbiota-derived metabolites and tissue tight junctions in rats under chronic sleep deprivation. Food Funct. 2022, 13, 2662–2680. [Google Scholar] [CrossRef]
  190. Falkenstein, M.; Simon, M.-C.; Mantri, A.; Weber, B.; Koban, L.; Plassmann, H. Impact of the gut microbiome composition on social decision-making. PNAS Nexus 2024, 3, pgae166. [Google Scholar] [CrossRef]
  191. Zhang, Q.; Chen, B.; Zhang, J.; Dong, J.; Ma, J.; Zhang, Y.; Jin, K.; Lu, J. Effect of prebiotics, probiotics, synbiotics on depression: Results from a meta-analysis. BMC Psychiatry 2023, 23, 477. [Google Scholar] [CrossRef]
  192. Zhu, R.; Fang, Y.; Li, H.; Liu, Y.; Wei, J.; Zhang, S.; Wang, L.; Fan, R.; Wang, L.; Li, S.; et al. Psychobiotic Lactobacillus plantarum JYLP-326 relieves anxiety, depression, and insomnia symptoms in test anxious college via modulating the gut microbiota and its metabolism. Front. Immunol. 2023, 14, 1158137. [Google Scholar] [CrossRef]
  193. Steenbergen, L.; Sellaro, R.; van Hemert, S.; Bosch, J.A.; Colzato, L.S. A randomized controlled trial to test the effect of multispecies probiotics on cognitive reactivity to sad mood. Brain Behav. Immun. 2015, 48, 258–264. [Google Scholar] [CrossRef]
  194. Matis, L.; Alexandru, B.A.; Fodor, R.; Daina, L.G.; Ghitea, T.C.; Vlad, S. Effect of Probiotic Therapy on Neuropsychiatric Manifestations in Children with Multiple Neurotransmitter Disorders: A Study. Biomedicines 2023, 11, 2643. [Google Scholar] [CrossRef]
  195. Arteaga-Henriquez, G.; Rosales-Ortiz, S.K.; Arias-Vasquez, A.; Bitter, I.; Ginsberg, Y.; Ibanez-Jimenez, P.; Kilencz, T.; Lavebratt, C.; Matura, S.; Reif, A.; et al. Treating impulsivity with probiotics in adults (PROBIA): Study protocol of a multicenter, double-blind, randomized, placebo-controlled trial. Trials 2020, 21, 161. [Google Scholar] [CrossRef] [PubMed]
  196. Ueda, E.; Matsunaga, M.; Fujihara, H.; Kajiwara, T.; Takeda, A.K.; Watanabe, S.; Hagihara, K.; Myowa, M. Temperament in early childhood is associated with gut microbiota composition and diversity. Dev. Psychobiol. 2024, 66, e22542. [Google Scholar] [CrossRef] [PubMed]
  197. Duan, Y.; Wu, X.; Yang, Y.; Gu, L.; Liu, L.; Yang, Y.; Zhou, J.; Wu, C.; Jin, F. Marked shifts in gut microbial structure and neurotransmitter metabolism in fresh inmates revealed a close link between gut microbiota and mental health: A case-controlled study. Int. J. Clin. Health Psychol. 2022, 22, 100323. [Google Scholar] [CrossRef] [PubMed]
  198. Prescott, S.L.; Logan, A.C.; LaFata, E.M.; Naik, A.; Nelson, D.H.; Robinson, M.B.; Soble, L. Crime and Nourishment: A Narrative Review Examining Ultra-Processed Foods, Brain, and Behavior. Dietetics 2024, 3, 318–345. [Google Scholar] [CrossRef]
  199. LaFata, E.M.; Allison, K.C.; Audrain-McGovern, J.; Forman, E.M. Ultra-Processed Food Addiction: A Research Update. Curr. Obes. Rep. 2024, 13, 214–223. [Google Scholar] [CrossRef] [PubMed]
  200. Murray, L.; Shaw, D.S.; Forbes, E.E.; Hyde, L.W. Reward-related neural correlates of antisocial behavior and callous–unemotional traits in young men. Biol. Psychiatry Cogn. Neurosci. Neuroimaging 2017, 2, 346–354. [Google Scholar] [CrossRef]
  201. Mesas, A.E.; de Andrade, S.M.; Melanda, F.N.; López-Gil, J.F.; Beneit, N.; Martínez-Vizcaíno, V.; Jiménez-López, E. Is Violence Victimization Associated with the Consumption of Ultra-Processed Food? A Population-Based Study with 96 K Adolescent Students Exploring the Mediating Role of Psychoactive Substance Use. Int. J. Ment. Health Addict. 2024, 1–7. [Google Scholar] [CrossRef]
  202. Dallman, M.F.; Akana, S.F.; Laugero, K.D.; Gomez, F.; Manalo, S.; Bell, M.; Bhatnagar, S. A spoonful of sugar: Feedback signals of energy stores and corticosterone regulate responses to chronic stress. Physiol. Behav. 2003, 79, 3–12. [Google Scholar] [CrossRef]
  203. Mesas, A.E.; Girotto, E.; Rodrigues, R.; Martínez-Vizcaíno, V.; Jiménez-López, E.; López-Gil, J.F. Ultra-processed food consumption is associated with alcoholic beverage drinking, tobacco smoking, and illicit drug use in adolescents: A nationwide population-based study. Int. J. Ment. Health Addict. 2023, 1–24. [Google Scholar]
  204. Samulėnaitė, S.; García-Blanco, A.; Mayneris-Perxachs, J.; Domingo-Rodríguez, L.; Cabana-Domínguez, J.; Fernàndez-Castillo, N.; Gago-García, E.; Pineda-Cirera, L.; Burokas, A.; Espinosa-Carrasco, J. Gut microbiota signatures of vulnerability to food addiction in mice and humans. Gut, 2024; ahead of print. [Google Scholar] [CrossRef]
  205. LaFata, E.M.; Worwag, K.; Derrigo, K.; Hessler, C.; Allison, K.C.; Juarascio, A.S.; Gearhardt, A.N. Development of the Food Addiction Symptom Inventory: The first clinical interview to assess ultra-processed food addiction. Psychol. Assess. 2024; ahead of print. [Google Scholar] [CrossRef] [PubMed]
  206. Basile, A.; Ruiz-Tejada, A.; Mohr, A.; Morales, A.; Hjelm, E.; Brand-Miller, J.; Atkinson, F.; Sweazea, K. Food Processing According to the NOVA Classification is Not Associated with Glycemic Index and Glycemic Load: Results from an Analysis of 1995 Food Items. Am. J. Clin. Nutr. 2024; ahead of print. [Google Scholar] [CrossRef] [PubMed]
  207. Bushman, B.J.; DeWall, C.N.; Pond, R.S., Jr.; Hanus, M.D. Low glucose relates to greater aggression in married couples. Proc. Natl. Acad. Sci. 2014, 111, 6254–6257. [Google Scholar] [CrossRef] [PubMed]
  208. Strang, S.; Hoeber, C.; Uhl, O.; Koletzko, B.; Münte, T.F.; Lehnert, H.; Dolan, R.J.; Schmid, S.M.; Park, S.Q. Impact of nutrition on social decision making. Proc. Natl. Acad. Sci. USA 2017, 114, 6510–6514. [Google Scholar] [CrossRef]
  209. Lucius, K. “Brain Fog”: Exploring a Symptom Commonly Encountered in Clinical Practice. Altern. Complement. Ther. 2021, 27, 23–30. [Google Scholar] [CrossRef]
  210. Becker-Blease, K.; Freyd, J.J. Dissociation and memory for perpetration among convicted sex offenders. J. Trauma Dissociation 2007, 8, 69–80. [Google Scholar] [CrossRef]
  211. Hopwood, J.; Snell, H. Amnesia in relation to crime. J. Ment. Sci. 1933, 79, 27–41. [Google Scholar] [CrossRef]
  212. Geng, F.; Lu, H.; Zhang, Y.; Zhan, N.; Zhang, L.; Liu, M. Dissociative depression and its related clinical and psychological characteristics among Chinese prisoners: A latent class analysis. Curr. Psychol. 2023, 42, 15070–15079. [Google Scholar] [CrossRef]
  213. Debowska, A.; Boduszek, D.; Ochman, M.; Hrapkowicz, T.; Gaweda, M.; Pondel, A.; Horeczy, B. Brain Fog Scale (BFS): Scale development and validation. Pers. Indiv. Differ. 2024, 216, 112427. [Google Scholar] [CrossRef]
  214. Jehangir, A.; Tetangco, E.P.; Ghafoor, A.; Yan, Y.; Laufer, A.; Karunaratne, T.; Inman, B.; Sharma, A.; Rao, S. S520 Development and Validation of Brain Fog Questionnaire in Patients and Healthy Volunteers. Am. J. Gastroenterol. 2022, 117, e368. [Google Scholar] [CrossRef]
  215. Rao, S.S.; Rehman, A.; Yu, S.; De Andino, N.M. Brain fogginess, gas and bloating: A link between SIBO, probiotics and metabolic acidosis. Clin. Transl. Gastroenterol. 2018, 9, e162. [Google Scholar] [CrossRef] [PubMed]
  216. Sheedy, J.R.; Wettenhall, R.E.; Scanlon, D.; Gooley, P.R.; Lewis, D.P.; Mcgregor, N.; Stapleton, D.I.; Butt, H.L.; De Meirleir, K.L. Increased d-lactic acid intestinal bacteria in patients with chronic fatigue syndrome. In Vivo 2009, 23, 621–628. [Google Scholar]
  217. George, D.T.; Hibbeln, J.R.; Ragan, P.W.; Umhau, J.C.; Phillips, M.J.; Doty, L.; Hommer, D.; Rawlings, R.R. Lactate-induced rage and panic in a select group of subjects who perpetrate acts of domestic violence. Biol. Psychiatry 2000, 47, 804–812. [Google Scholar] [CrossRef]
  218. Furlan, F.; Hoshino, K. Fighting by sleep-deprived rats as a possible manifestation of panic: Effects of sodium lactate. Braz. J. Med. Biol. Res. 2001, 34, 359–366. [Google Scholar] [CrossRef] [PubMed]
  219. Petersen, C. D-lactic acidosis. Nutr. Clin. Pract. 2005, 20, 634–645. [Google Scholar] [CrossRef]
  220. Hanstock, T.L.; Clayton, E.H.; Li, K.M.; Mallet, P.E. Anxiety and aggression associated with the fermentation of carbohydrates in the hindgut of rats. Physiol. Behav. 2004, 82, 357–368. [Google Scholar] [CrossRef] [PubMed]
  221. Kowlgi, N.G.; Chhabra, L. D-lactic acidosis: An underrecognized complication of short bowel syndrome. Gastroenterol. Res. Pract. 2015, 2015, 476215. [Google Scholar] [CrossRef]
  222. Lefèvre, C.R.; Turban, A.; Paz, D.L.; Penven, M.; René, C.; Langlois, B.; Pawlowski, M.; Collet, N.; Piau, C.; Cattoir, V. Early detection of plasma D-lactate: Toward a new highly-specific biomarker of bacteraemia? Heliyon 2023, 9, e16466. [Google Scholar] [CrossRef]
  223. Hecht, A.L.; Harling, L.C.; Friedman, E.S.; Tanes, C.; Lee, J.; Firrman, J.; Hao, F.; Tu, V.; Liu, L.; Patterson, A.D. Dietary carbohydrates regulate intestinal colonization and dissemination of Klebsiella pneumoniae. J. Clin. Investig. 2024, 134, e174726. [Google Scholar] [CrossRef]
  224. Zhu, Y.; Mierau, J.O.; Riphagen, I.J.; Heiner-Fokkema, M.R.; Dekker, L.H.; Navis, G.J.; Bakker, S.J.L. Types of fish consumption differ across socioeconomic strata and impact differently on plasma fish-based omega-3 fatty acids: A cross-sectional study. Eur. J. Nutr. 2024, 63, 435–443. [Google Scholar] [CrossRef]
  225. Aljahdali, A.A.; Rossato, S.L.; Baylin, A. Ultra-processed foods consumption among a USA representative sample of middle-older adults: A cross-sectional analysis. Br. J. Nutr. 2024, 131, 1461–1472. [Google Scholar] [CrossRef] [PubMed]
  226. Maury, A.A.; Holton, K. The Low Glutamate Diet Improves Sleep and Cognition in Veterans With Gulf War Illness. Curr. Develop. Nutr. 2024, 8, 103202. [Google Scholar] [CrossRef]
  227. Gbadamosi, I.; Yawson, E.O.; Akesinro, J.; Adeleke, O.; Tokunbo, O.; Bamisi, O.; Ibrahim-Abdulkareem, R.; Awoniran, P.; Gbadamosi, R.; Lambe, E.; et al. Vitamin D attenuates monosodium glutamate-induced behavioural anomalies, metabolic dysregulation, cholinergic impairment, oxidative stress, and astrogliosis in rats. Neurotoxicology 2024, 103, 297–309. [Google Scholar] [CrossRef] [PubMed]
  228. Wiss, D.A.; LaFata, E.M. Ultra-Processed Foods and Mental Health: Where Do Eating Disorders Fit into the Puzzle? Nutrients 2024, 16, 1955. [Google Scholar] [CrossRef]
  229. Fazzino, T.L.; Jun, D.; Chollet-Hinton, L.; Bjorlie, K. US tobacco companies selectively disseminated hyper-palatable foods into the US food system: Empirical evidence and current implications. Addiction 2024, 119, 62–71. [Google Scholar] [CrossRef]
  230. de Noronha, S.I.R.; de Moraes, L.A.G.; Hassell, J.E., Jr.; Stamper, C.E.; Arnold, M.R.; Heinze, J.D.; Foxx, C.L.; Lieb, M.M.; Cler, K.E.; Karns, B.L. High-fat diet, microbiome-gut-brain axis signaling, and anxiety-like behavior in male rats. Biol. Res. 2024, 57, 23. [Google Scholar] [CrossRef]
  231. Reuter, M.; Zamoscik, V.; Plieger, T.; Bravo, R.; Ugartemendia, L.; Rodriguez, A.B.; Kirsch, P. Tryptophan-rich diet is negatively associated with depression and positively linked to social cognition. Nutr. Res. 2021, 85, 14–20. [Google Scholar] [CrossRef]
  232. Chojnacki, C.; Gąsiorowska, A.; Popławski, T.; Konrad, P.; Chojnacki, M.; Fila, M.; Blasiak, J. Beneficial effect of increased tryptophan intake on its metabolism and mental state of the elderly. Nutrients 2023, 15, 847. [Google Scholar] [CrossRef]
  233. Zamoscik, V.; Schmidt, S.N.L.; Bravo, R.; Ugartemendia, L.; Plieger, T.; Rodríguez, A.B.; Reuter, M.; Kirsch, P. Tryptophan-enriched diet or 5-hydroxytryptophan supplementation given in a randomized controlled trial impacts social cognition on a neural and behavioral level. Sci. Rep. 2021, 11, 21637. [Google Scholar] [CrossRef]
  234. Young, S.N. The effect of raising and lowering tryptophan levels on human mood and social behaviour. Philos. Trans. R. Soc. B Biol. Sci. 2013, 368, 20110375. [Google Scholar] [CrossRef]
  235. Duquenne, P.; Capperella, J.; Fezeu, L.K.; Srour, B.; Benasi, G.; Hercberg, S.; Touvier, M.; Andreeva, V.A.; St-Onge, M.-P. The association between ultra-processed food consumption and chronic insomnia in the NutriNet-Santé Study. J. Acad. Nutr. Diet. 2024, 124, 1109–1117.e2. [Google Scholar] [CrossRef] [PubMed]
  236. Zhu, C.; Sawrey-Kubicek, L.; Beals, E.; Rhodes, C.H.; Houts, H.E.; Sacchi, R.; Zivkovic, A.M. Human gut microbiome composition and tryptophan metabolites were changed differently by fast food and Mediterranean diet in 4 days: A pilot study. Nutr. Res. 2020, 77, 62–72. [Google Scholar] [CrossRef] [PubMed]
  237. Salzer, H.M. Relative hypoglycemia as a cause of neuropsychiatric illness. J. Natl. Med. Assoc. 1966, 58, 12. [Google Scholar]
  238. Wilder, J. Sugar metabolism and its relation to criminology. In Handbook of Correctional Psychology; Lindner, R.M., Seliger, R.V., Eds.; Philosophical Library: New York, NY, USA, 1947; pp. 98–129. [Google Scholar]
  239. Donohoe, R.T.; Benton, D. Blood glucose control and aggressiveness in females. Pers. Indiv. Differ. 1999, 26, 905–911. [Google Scholar] [CrossRef]
  240. Benton, D.; Kumari, N.; Brain, P.F. Mild hypoglycaemia and questionnaire measures of aggression. Biol. Psychol. 1982, 14, 129–135. [Google Scholar] [CrossRef] [PubMed]
  241. Virkkunen, M. Reactive Hypoglyceinic Tendency among Habitually Violent Offenders: A Further Study by Means of the Glucose Tolerance Test. Neuropsychobiology 1982, 8, 35–40. [Google Scholar] [CrossRef]
  242. Virkkunen, M. Insulin-Secretion during the Glucose-Tolerance Test among Habitually Violent and Impulsive Offenders. Aggress. Behav. 1986, 12, 303–310. [Google Scholar] [CrossRef]
  243. Virkkunen, M. Insulin secretion during the glucose tolerance test in antisocial personality. Br. J. Psychiatry 1983, 142, 598–604. [Google Scholar] [CrossRef] [PubMed]
  244. Virkkunen, M. Reactive hypoglycemic tendency among arsonists. Acta Psychiatr. Scand. 1984, 69, 445–452. [Google Scholar] [CrossRef]
  245. Pfundmair, M.; DeWall, C.N.; Fries, V.; Geiger, B.; Kramer, T.; Krug, S.; Frey, D.; Aydin, N. Sugar or spice: Using I3 metatheory to understand how and why glucose reduces rejection-related aggression. Aggress. Behav. 2015, 41, 537–543. [Google Scholar] [CrossRef]
  246. Xu, H.; Bègue, L.; Sauve, L.; Bushman, B.J. Sweetened blood sweetens behavior. Ego depletion, glucose, guilt, and prosocial behavior. Appetite 2014, 81, 8–11. [Google Scholar] [CrossRef] [PubMed]
  247. DeWall, C.N.; Deckman, T.; Gailliot, M.T.; Bushman, B.J. Sweetened blood cools hot tempers: Physiological self-control and aggression. Aggress. Behav. 2011, 37, 73–80. [Google Scholar] [CrossRef]
  248. Virkkunen, M.; Rawlings, R.; Tokola, R.; Poland, R.E.; Guidotti, A.; Nemeroff, C.; Bissette, G.; Kalogeras, K.; Karonen, S.-L.; Linnoila, M. CSF biochemistries, glucose metabolism, and diurnal activity rhythms in alcoholic, violent offenders, fire setters, and healthy volunteers. Arch. Gen. Psychiatry 1994, 51, 20–27. [Google Scholar] [CrossRef] [PubMed]
  249. Mishra, P. Neuroscientific Paradigms and their Implications for Jurisprudential Practice: A Comparative Analysis. Athens J. LAW 2024, 10, 317. [Google Scholar] [CrossRef]
  250. Borbón, D. Free will, quarantines, and moral enhancements: Neuroabolitionism as an alternative to criminal law. Front. Sociol. 2024, 9, 1395986. [Google Scholar] [CrossRef] [PubMed]
  251. Schneider, E.; O’Riordan, K.J.; Clarke, G.; Cryan, J.F. Feeding gut microbes to nourish the brain: Unravelling the diet–microbiota–gut–brain axis. Nat. Metab. 2024, 6, 1454–1478. [Google Scholar] [CrossRef]
  252. Logan, A.C.; Schoenthaler, S.J. Nutrition, Behavior, and the Criminal Justice System: What Took so Long? An Interview with Dr. Stephen J. Schoenthaler. Challenges 2023, 14, 37. [Google Scholar] [CrossRef]
  253. Wang, F.L.; Bountress, K.E.; Lemery-Chalfant, K.; Wilson, M.N.; Shaw, D.S. A polygenic risk score enhances risk prediction for adolescents’ antisocial behavior over the combined effect of 22 extra-familial, familial, and individual risk factors in the context of the family check-up. Prev. Sci. 2023, 24, 739–751. [Google Scholar] [CrossRef]
  254. Prescott, S.L.; Logan, A.C. Each meal matters in the exposome: Biological and community considerations in fast-food-socioeconomic associations. Econ. Hum. Biol. 2017, 27, 328–335. [Google Scholar] [CrossRef]
  255. DeAngelis, T. Continuing Education: Nutrition’s role in mental health. Monit. Psychol. 2023, 54, 36–41. [Google Scholar]
Figure 1. Recent cases involving the dismissal of driving while intoxicated charges after lab-proven internal alcohol production (i.e., “auto-brewery” via food and microbe interactions, without alcohol consumption) demonstrate that the courts are opening to the legalome and variants of the Twinkie Defense (used with permission of Susan L. Prescott).
Figure 1. Recent cases involving the dismissal of driving while intoxicated charges after lab-proven internal alcohol production (i.e., “auto-brewery” via food and microbe interactions, without alcohol consumption) demonstrate that the courts are opening to the legalome and variants of the Twinkie Defense (used with permission of Susan L. Prescott).
Neurosci 05 00028 g001
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Prescott, S.L.; Holton, K.F.; Lowry, C.A.; Nicholson, J.J.; Logan, A.C. The Intersection of Ultra-Processed Foods, Neuropsychiatric Disorders, and Neurolaw: Implications for Criminal Justice. NeuroSci 2024, 5, 354-377. https://doi.org/10.3390/neurosci5030028

AMA Style

Prescott SL, Holton KF, Lowry CA, Nicholson JJ, Logan AC. The Intersection of Ultra-Processed Foods, Neuropsychiatric Disorders, and Neurolaw: Implications for Criminal Justice. NeuroSci. 2024; 5(3):354-377. https://doi.org/10.3390/neurosci5030028

Chicago/Turabian Style

Prescott, Susan L., Kathleen F. Holton, Christopher A. Lowry, Jeffrey J. Nicholson, and Alan C. Logan. 2024. "The Intersection of Ultra-Processed Foods, Neuropsychiatric Disorders, and Neurolaw: Implications for Criminal Justice" NeuroSci 5, no. 3: 354-377. https://doi.org/10.3390/neurosci5030028

Article Metrics

Back to TopTop