Your cart

Your cart is empty

Mycotoxin Bioremediation by the Gut Microbiota

Mycotoxin Bioremediation by the Gut Microbiota

Mycotoxins are increasingly becoming a concern as the Environmental Protection Agency reports up to 85% of buildings have been water-damaged [1]. Your patients may not know it, but if they have obscure symptoms and conditions, mold toxins could be a contributor. These symptoms can include:

  • Brain fog

  • Suboptimal immune function

  • Worsened allergies

  • Strange neurological symptoms

  • Unexplained weight gain and inability to keep lost weight off

When it comes to mold toxin responses, it truly depends on individual genetics, metabolism, gut flora, toxic load, and even prior infections—all factors that determine individual bioremediation capacity. Some individuals may experience zero symptoms, while others become very sick, bedridden, and sensitive to the slightest stimuli. 

Currently, there are:

  • Blood and urine tests for mycotoxin levels.

  • Genetic tests for mycotoxin response, such as Human Leukocyte Antigens (HLAs).

  • Fungal IgE and IgA tests.

  • Specialty inflammatory markers, such as MMP9, MSH, FSH, ADH, and TGF-beta. 

With these tests, you can diagnose mold toxicity with specific types of mold and which binders work best for each patient. However, no test can precisely determine individual biochemical and microbial responses to their mold toxins. 

In this article, we'll cover the role of the gut microbiota and how our new test, Elie, helps you better understand these aspects of health so you can create a more personalized mold detox protocol.

What Are Mycotoxins and How Do They Affect Your Health?

Mycotoxins, such as ochratoxin A and aflatoxins, are toxic secondary metabolites produced by fungi, commonly found in mold-contaminated environments. Also, common fermented and dried foods like grains, coffee, nuts, dried fruits, and cacao may contain some mycotoxins.

These compounds are highly stable, resistant to heat, and difficult to eliminate, which means they persist in the body long after exposure.

Some individuals may clear them efficiently and have no symptoms, while others experience significant health consequences. Certain mycotoxins are also known carcinogens and have been linked to liver and kidney toxicity [2].

One of the most overlooked consequences of mycotoxin exposure is unexplained weight gain and difficulty losing weight. Since these toxins are lipophilic (fat-loving), the body stores them in fat cells as a protective mechanism. This can lead to metabolic dysfunction, making it harder to mobilize fat stores during weight loss efforts.

Currently, mycotoxin exposure is primarily assessed through blood and urine tests, but these only measure circulating or excreted toxins at a given moment. They don’t provide insight into how well the body is actively managing and neutralizing these toxins. 

This is where the gut microbiota plays a critical role. Certain gut bacteria can bind, degrade, and biotransform mycotoxins, reducing their toxicity and systemic burden. Given the increasing prevalence of mycotoxin-related illnesses, understanding a patient’s gut microbiome is crucial in developing targeted detoxification strategies. 

With Elie’s advanced microbiome testing, clinicians can assess their patients’ microbial profiles to determine whether they have the necessary bacterial strains to mitigate mycotoxin toxicity effectively. 

First, let’s understand how these mycotoxins affect gut health and flora.

Why does the gut microbiome matter for mold detox?

  • The gut microbiota are your back pocket of genes that defend against mycotoxins–they have perhaps 150 times more genes than humans do [3]. Some beneficial bacteria help neutralize and break down mycotoxins, preventing them from entering circulation.

  • If gut bacteria are damaged by mycotoxins, leaky gut and dysbiosis (microbial imbalances) can develop, making it harder to detox and increasing systemic inflammation.

  • The gut microbiota can easily change in response to the right interventions.

  • Understanding your gut microbiome’s ability to bioremediate (break down) mycotoxins can help guide a more precise mold detox strategy.

How Do Mycotoxins Affect Gut Health and Gut Flora?

Mycotoxins can enter the body through contaminated food and water, and the gastrointestinal tract is the first line of exposure. If you inhale mold toxins from your environment, you’ll eventually swallow the postnasal drip, exposing your gut to the toxins. 

Mycotoxin-Induced Disruption of Gut Microbiota

Many mycotoxins exhibit antimicrobial properties that selectively eliminate beneficial bacteria while allowing pathogenic species to flourish.

Chronic exposure can shift the balance of key bacterial phyla, reducing Firmicutes and Bacteroidetes while increasing Proteobacteria, a phylum linked to gut inflammation and disease [4].

Gut Permeability and Immune Dysregulation

Mycotoxins like ochratoxin A (OTA) and aflatoxin B1 (AFB1) damage intestinal epithelial cells, weakening tight junctions and increasing permeability. This allows toxins, pathogens, and microbial byproducts such as lipopolysaccharides to reach circulation, triggering systemic inflammation. The immune response to this exposure can worsen gut dysbiosis, further worsening the cycle of gut dysfunction.

Secondary Metabolic Disruptions

Mycotoxins can alter gut microbial metabolism, affecting the production of key bioactive compounds such as short-chain fatty acids (SCFAs) and sphingolipids. These molecules play vital roles in immune regulation and metabolic health. 

A reduction in SCFAs like butyrate can impair gut barrier function, decreasing protection against inflammatory disorders like inflammatory bowel disease (IBD) and increasing susceptibility to cancer [5].

Altered sphingolipid metabolism has been linked to metabolic disorders such as [6]:

  • Obesity

  • Type 2 diabetes

  • Cardiovascular disease

  • Liver disease

Specific Microbiome Exposures to Each Type of Mycotoxin

A meta-analysis of nearly 100 studies analyzed the effects of different mycotoxins on gut function, identifying the following key mycotoxins [7]:

Aflatoxins (AFB1, AFB2, AFM1)

Produced by fungi (Aspergillus spp.) [8]. Aflatoxin exposures can lead to:

  • Increased intestinal permeability.

  • Microbiome alterations, which reduce beneficial Lactobacillus and Bifidobacterium populations, leading to dysbiosis.

  • Oxidative stress and inflammation by elevating IL-6, TNF-α, and reactive oxygen species (ROS).

Ochratoxin A (OTA)

Produced by Aspergillus spp. and Penicillin v. [9]. These can lead to:

  • Mucosal damage: Causes intestinal villi wasting (atrophy) and crypt overgrowth (hyperplasia), impairing nutrient absorption.

  • Microbial shift: Increases pathogenic Enterobacteriaceae while decreasing beneficial Firmicutes.

  • Decreased IgA production: Increases susceptibility to toxins and gut infections.

Fumonisins (FB1, FB2)

Produced by Fusarium spp. [10]. It can lead to:

  • Disrupts sphingolipid metabolism, which alters the intestinal cell membrane and reduces gut barrier function.

  • Increases pathogenic bacteria like Escherichia coli and Clostridium spp., which contribute to gut inflammation.

  • Affects neuroinflammation and neurotransmitter functions, changing the Gut-Brain Axis.

Zearalenone (ZEA)

Produced by Fusarium [11]. It can lead to:

  • Hormone disruption: ZEA mimics estrogen and affect microbial metabolism of bile acids and hormones.

  • Microbial imbalance by increasing opportunistic fungi and yeasts while reducing good bacteria.

  • Increased pro-inflammatory cytokines (IL-1β, IL-6, TNF-α).

Deoxynivalenol (DON, “Vomitoxin”)

Produced by Fusarium [12]. It can lead to:

  • Gut hyperpermeability by triggering tight junction breakdown, leading to increased toxin absorption.

  • Reduces beneficial bacteria involved in SCFA production.

  • Nausea and vomiting by affecting gut serotonin levels and changing gut motility and sickness response.

Patulin

Produced by Aspergillus spp. and Penicillin spp.[13]. It can lead to:

  • Intestinal cell damage through apoptosis, compromising gut lining integrity.

  • Dysbiosis through reducing Lactobacillus spp., leading to increased opportunistic infections.

  • Weakened immunity by reducing mucin production, making the gut lining more vulnerable to pathogens.

In short, mycotoxins cause dysbiosis and intestinal permeability, while weakening certain immune defenses. If you have a patient with mycotoxins, you’d want to help them remove them as quickly as possible. However, some mold patients are very sensitive because mold toxins can inhibit some detoxification pathways, so detoxes often have to be slow, methodical, and individualized.

How Are Mycotoxins Eliminated From the Body?

The human body employs a three-phase detoxification system to process and eliminate harmful substances, including mycotoxins [14],[15]. These toxins, once absorbed through the gut, mostly undergo detoxification in the liver before being excreted through bile, urine, and feces.

The gut microbiota assists this process by binding and metabolizing some mycotoxins to prevent their systemic absorption. It also supports various liver detoxification pathways.

If your microbiome is imbalanced, mycotoxins can recirculate and cause persistent symptoms. The Elie test helps assess your microbial detox capacity to optimize mycotoxin clearance. I may also help avoid things that can increase the mycotoxin reactivation or reabsorption.

Phase 1: Activation and Biotransformation

Phase 1 detoxification is the initial step in metabolizing mycotoxins, transforming them into more water-soluble forms to facilitate excretion. 

This phase primarily involves a group of enzymes known as cytochrome P450 enzymes (CYP450), which catalyze oxidation, reduction, and hydrolysis reactions [16].

Key Enzymes and Mechanisms

  • CYP450 enzymes add oxygen (oxidize) to mycotoxins, making them more reactive.

  • Hydroxylation and dealkylation reactions prepare mycotoxins for further modification.

This process, while necessary, can produce reactive intermediates that may be even more toxic than the original mycotoxin if not efficiently processed in Phase 2 [17].

Gut Microbiota’s Role

Certain gut bacteria, like Lactobacillus and Bifidobacterium species, can degrade mycotoxins directly, preventing them from reaching the liver [18],[19].

Some microbes transform toxic mycotoxins into less harmful metabolites, while others bind toxins to their cell walls, reducing their absorption.

Phase 2: Conjugation and Neutralization

Phase 2 detoxification involves conjugation, where the liver attaches molecules to mycotoxin metabolites to render them less reactive and more water-soluble for excretion. This step is critical for neutralizing the toxic intermediates produced in Phase 1.

Key Enzymes and Conjugation Pathways

  • Glutathione S-transferases (GST): Glutathione binds to toxic intermediates, neutralizing oxidative stress from mycotoxin exposure [20].

  • Sulfotransferases (SULTs): Attach sulfate groups to mycotoxins, making them easier to excrete [21].

  • UDP-glucuronosyltransferases (UGTs): Facilitate glucuronidation, making mycotoxins more water-soluble for biliary and urinary excretion [17].

Gut Microbiota’s Role

Certain gut microbes can enzymatically modify mycotoxins. In some cases, this detoxifies the mycotoxins. In others, it bioactivates the mycotoxins, making the toxins more harmful.

The gut bacteria can convert deoxynivalenol (DON) into a less toxic metabolite (DOM-1). Some microbes bind mycotoxins to their cell walls, limiting absorption and systemic exposure.

However, masked mycotoxins (glucoside-conjugated forms) may be hydrolyzed into their active, more toxic forms [22].

The gut microbiota contributes by regulating the availability of conjugation building blocks, such as sulfate, glucuronide, and glutathione [23].

Some gut microbes break down mycotoxins before they even reach the liver, reducing the burden on detoxification pathways.

Dysbiosis can impair Phase 2 detoxification, leading to poor elimination and increased toxin recirculation.

Phase 3: Transport and Excretion

Once neutralized, mycotoxins must be efficiently excreted from the body. Phase 3 detoxification involves active transport proteins that shuttle mycotoxins and their conjugates from liver cells into bile, urine, or feces.

Key Transporters and Excretion Pathways
  • Multidrug resistance proteins (MRPs) transport mycotoxins from liver cells into bile, where they enter the digestive tract and are excreted in feces [24].

  • Organic anion transporters (OATs) facilitate urinary elimination [25].

Gut Microbiota’s Role

The gut flora assists by metabolizing and binding toxins excreted in bile, preventing reabsorption.

A healthy gut microbiome prevents enterohepatic recirculation, where mycotoxins excreted in bile are reabsorbed in the intestines.

Bacterial strains, such as Lactobacillus rhamnosus and Bifidobacterium bifidum, bind and degrade mycotoxins in the gut, further aiding in elimination.

Cases of dysbiosis (imbalanced gut flora) can lead to the reabsorption of mycotoxins, prolonging toxicity.

How Does Microbiome Testing Help Mold Patients?

Since the gut flora plays a critical role in all three detoxification phases, understanding an individual’s microbiome composition can offer insights into their detoxification efficiency. 

The Elie test provides a detailed analysis of gut microbial populations, allowing clinicians to identify gaps in mycotoxin detoxification capacity and implement targeted interventions such as:

  • Supporting beneficial bacteria that aid in mycotoxin degradation.

  • Addressing dysbiosis that may impair elimination and cause enterohepatic recirculation.

  • Enhancing liver detox pathways by optimizing glutathione, sulfur, and other conjugation building blocks.

Common Complications With Mycotoxin Detox

Mycotoxin detoxification is not always straightforward. Many patients, particularly those with significant mold exposure, experience severe reactions when attempting to eliminate mycotoxins from their bodies. 

These reactions often stem from:

  • Disrupted detoxification process

  • Heightened sensitivity

  • Excessive toxin mobilization

Below, we discuss the most common complications and why a gradual, personalized approach is necessary for a safe and effective detox.

Jarisch-Herxheimer (JHR) Reactions: Too Many Toxins, Too Fast

One of the most common pitfalls in mycotoxin detoxification is an intense JHR reaction—a temporary worsening of symptoms caused by the rapid mobilization of toxins. As mycotoxins are released from fat cells or other storage sites, the body may struggle to excrete them efficiently, leading to a toxic backlog that triggers inflammation and immune system activation [26].

Symptoms of a JHR may include:

  • Brain fog

  • Fatigue

  • Joint and muscle pain

  • Flu-like symptoms

  • Nausea and vomiting

  • Flushing

  • Anxiety or mood swings

  • Headaches and dizziness

What Causes It?

Detox binders, such as activated charcoal or cholestyramine, can pull toxins out of fat cells into circulation. If elimination and detoxification pathways aren’t optimized, the patient may experience toxicity symptoms. This is why titration is important for sensitive patients.

A sluggish liver, poor bile flow, or gut dysbiosis can lead to recirculation of mycotoxins, worsening symptoms instead of eliminating toxins.

Impaired Phase 2 Detoxification: The “Bottleneck” Effect

A critical mistake in mycotoxin detox protocols is supporting Phase 1 detox without addressing Phase 2. This imbalance can lead to the buildup of harmful intermediate metabolites, which are often more toxic and reactive than the original mycotoxin.

Although scientific data on interphase transitions is limited, we know that Phase 1 detox enzymes break down mycotoxins, making them more water-soluble. However, some byproducts of Phase 1 detox are more reactive and damaging to cells.

If Phase 2 detox pathways (such as glucuronidation, sulfation, and glutathione conjugation) aren’t working efficiently, these intermediates accumulate, causing oxidative stress and worsening symptoms.

Many mold patients have impaired Phase 2 detox due to genetic predispositions, chronic inflammation, or liver congestion. Mold toxins can also impair Phase 2 detoxification.

Signs of Phase 2 impairment may include:

  • Sensitivity to supplements or detox therapies

  • Worsening fatigue or neurological symptoms with detox attempts

  • Increased inflammation, headaches, or joint pain.

To prevent this, it’s crucial to ensure Phase 2 detoxification works before engaging in a more aggressive mold detox. 

A clinical trial evaluated the impact of zinc and vitamin C supplementation on oxidative stress and liver toxicity caused by Aflatoxin B1 (AFB1) exposure in 35 wheat millers. Liver enzymes, oxidative stress markers, and detoxification proteins (P53, zinc, and vitamin C) were measured before and after supplementation [27].

Supplementation resulted in:

  • Reduced liver damage as evidenced by significant decreases in liver enzymes (AST, ALP, GGT) and oxidative stress markers (MDA, GST).

  • Detoxification protein P53 levels decreased in more than half of the participants, though not to a statistically significant degree.

Zinc and vitamin C supplementation helped counteract AFB1-induced hepatotoxicity, likely by reducing oxidative stress and improving liver function. Further large-scale studies are needed to confirm these findings.

Histamine Intolerance and Mast Cell Activation Syndrome (MCAS)

Mast cells are immune cells found in tissues like the skin, lungs, and gut, where they play a key role in inflammatory and allergic responses. They release histamine and other mediators to fight infections and heal wounds, but they can also contribute to excessive inflammation.

MCAS is a disorder where mast cells inappropriately release excessive chemicals, leading to systemic symptoms. Triggers vary but include stress, infections, and environmental toxins. Due to the multisystemic symptoms, MCAS can be very difficult to correctly diagnose and manage.

Symptoms may include [28]:

  • Flushing

  • Shortness of breath

  • Hives

  • Diarrhea

  • Vomiting

  • Life-threatening anaphylaxis 

Some mold patients may instead develop histamine intolerance due to inflammation of the gut lining. Also, the mold toxins may reduce beneficial bacteria that degrade histamine. Inflammation from mold exposure can lead to excessive mast cell activation and histamine release [29]. Poor liver detoxification impairs histamine clearance, worsening symptoms.

The following steps may minimize histamine reactions during detox:

  • Stabilize mast cells before detox (quercetin, vitamin C, DAO enzymes) [30].

  • Support gut flora to improve histamine breakdown (probiotics, prebiotics) [31].

  • Reduce high-histamine foods during detox (fermented foods, aged cheeses, alcohol).

The Importance of a Gradual, Holistic Detox Approach

Due to the potential complications with mold detoxes, mold patients tend to be tough cases. Therefore, mold detoxes have to be introduced gradually to prevent the worsening of symptoms. Here’s how to ensure a safe and effective detoxification process:

Step 1: Prepare the Body for Detox

  • For many patients, this step involves moving to a mold-free environment and cleaning up their diet, lifestyle, sleep, mindset, and addressing their traumas and nervous system.

  • Support liver function before mobilizing toxins.

  • Ensure bile flow is optimized with bitters and phosphatidylcholine.

  • Correct gut imbalances to prevent toxin recirculation.

  • Ensure sufficient water intake and regular bowel movements.

Step 2: Mobilize Toxins Slowly

  • Start with gentle binders (such as zeolite or bentonite clay) at low doses before gradually increasing to full doses with stronger options.

  • Use low-dose sauna therapy or lymphatic movement to encourage elimination.

Step 3: Enhance Elimination Pathways

  • Support kidney detoxification (hydration, electrolytes).

  • Use antioxidants, if tolerated, to neutralize reactive intermediates (glutathione, vitamin C, selenium).

How Elie and Aristotle Support Safe and Effective Mycotoxin Removal

Successfully eliminating mycotoxins from the body requires more than just binders and detox supplements—it demands an individualized approach that considers a patient’s gut microbiome health, detoxification capacity, and metabolic functions.

Everyone’s microbiome is unique and a key determinant of individual responses to mycotoxins. Elie and Aristotle provide data-driven insights that help clinicians personalize and optimize overall protocols for each patient. 

Aristotle: Assessing Toxin Load and Detoxification Capacity

Aristotle, a comprehensive metabolomic test, measures a patient’s toxic burden and detoxification efficiency.

Environmental Toxin Exposure Index

This index evaluates markers of environmental toxin exposure to determine toxin accumulation levels that may explain mycotoxin-related symptoms.

Liver Health and Detox Pathway Assessment

Since mycotoxin detox heavily depends on liver function, Aristotle can broadly analyze detoxification pathways to assess whether a patient can effectively process and eliminate toxins.

Aristotle can theoretically identify potential impairments in detox pathways that could lead to toxin buildup and worsening symptoms.

Oxidative Stress and Inflammation Markers

Aristotle detects chronic inflammation, overactive immune responses, and mitochondrial dysfunction, all of which can impact toxin clearance and overall health.

While Aristotle does not provide mold-specific protocols, it helps clinicians determine whether a patient can handle detoxification at a normal pace or if they require a more cautious, gradual approach to avoid overwhelming their system.

Elie: Understanding the Gut Microbiota’s Role in Mycotoxin Bioremediation

Mycotoxins can wreck the gut flora, but at the same time, some bacteria can degrade or activate mycotoxins. Elie can address these issues at the following levels:

Microbiome Analysis for Mycotoxin Degradation

Elie identifies beneficial bacterial strains that can neutralize and metabolize mycotoxins and helps determine if a patient needs specific probiotic or prebiotic interventions.

Dysbiosis Markers and Detox Readiness

Elie also assesses gut imbalances that may impair toxin elimination or lead to mycotoxin reabsorption. Identification of leaky gut markers that can increase systemic inflammation and detox side effects is also vital in this process.

Digital Twinning Protocol for Gut Optimization

While Elie does not provide specific mycotoxin detoxification protocols, it offers Digital Twinning recommendations for:

  • Targeted probiotic and prebiotic strategies to boost detox-friendly bacteria and pathways.

  • Precise dietary and supplemental recommendations for microbial balance. 

  • Dietary recommendations to enhance the gut’s detoxification capacities.

Integrating Elie and Aristotle for Personalized Mycotoxin Detox

Elie and Aristotle tests can be combined to create a powerful personalized plan to mycotoxin elimination: 

  1. Assess Detox Capacity (Aristotle)

    • Identify toxin exposure levels.

    • Evaluate liver detox function.

    • Determine oxidative stress and inflammation status.

  2. Assess Microbiome Health (Elie)

    • Identify gut flora’s mycotoxin degradation capacity.

    • Address dysbiosis 

    • Optimize microbial function with Digital Twinning.

  3. Personalize the Detox Strategy

    • Start with the whole body, especially gut and liver, support before mobilizing mycotoxins.

    • Introduce binders cautiously based on mycotoxin types (identified with another test), microbiome, and detox status.

    • Adjust probiotics, prebiotics, and dietary recommendations based on Elie’s insights.

Conclusion: The Future of Precision Mycotoxin Detox

Mycotoxins affect everyone differently: what works for one person may overwhelm another. For many challenging and complex cases, symptoms can also vary so much that they aren’t reliable indicators of how well a treatment is working. Therefore, having objective metabolomic and gut microbiome tests can provide a unique data-driven protocol and more accurately assess patient progress.

With Elie and Aristotle, you can:

  • Identify how your gut microbiome helps or hinders detox.

  • Assess liver detox efficiency and toxin load, identifying poor detoxes and where they need support to minimize Jarisch-Herxheimer reactions.

  • Fine-tune interventions for safe detoxifications.

With precise data-driven insights and tools like Digital Twinning, you can make mycotoxin detoxification safer and more effective.




1 US EPA. (2014) Summarized Data of the Building Assessment Survey and Evaluation Study

2 Ekwomadu, T., Mwanza, M. and Musekiwa, A. (2022) Mycotoxin-Linked Mutations and Cancer Risk: A Global Health Issue. Int J Environ Res Public Health 19 

3 Zhu, B., Wang, X. and Li, L. (2010) Human Gut Microbiome: The Second Genome of Human Body. Protein Cell 1, 718–725 

4 Rizzatti, G., Lopetuso, L. R., Gibiino, G., Binda, C. and Gasbarrini, A. (2017) Proteobacteria: A Common Factor in Human Diseases. Biomed Res Int 2017, 9351507 

5 Canani, R. B., Costanzo, M. D., Leone, L., Pedata, M., Meli, R. and Calignano, A. (2011) Potential Beneficial Effects of Butyrate in Intestinal and Extraintestinal Diseases. World J. Gastroenterol. 17, 1519–1528 

6 Green, C. D., Maceyka, M., Cowart, L. A. and Spiegel, S. (2021) Sphingolipids in Metabolic Disease: The Good, the Bad, and the Unknown. Cell Metab 33, 1293–1306 

7 Grenier, B. and Applegate, T. J. (2013) Modulation of Intestinal Functions Following Mycotoxin Ingestion: Meta-analysis of Published Experiments in Animals. Toxins (Basel) 5, 396–430 

8 (2015) Aflatoxins National Cancer Institute 

9 Bui-Klimke, T. R. and Wu, F. (2015) Ochratoxin A and Human Health Risk: A Review of the Evidence. Crit Rev Food Sci Nutr 55, 1860–1869 

10 Human Foods Program. (2024) Guidance for Industry: Fumonisin Levels in Human Foods and Animal Feeds. U.S. Food and Drug Administration, FDA 

11 Ropejko, K. and Twarużek, M. (2021) Zearalenone and Its Metabolites-General Overview, Occurrence, and Toxicity. Toxins (Basel) 13 

12 Sobrova, P., Adam, V., Vasatkova, A., Beklova, M., Zeman, L. and Kizek, R. (2010) Deoxynivalenol and its Toxicity. Interdiscip Toxicol 3, 94–99 

13 Zhong, L., Carere, J., Lu, Z., Lu, F. and Zhou, T. (2018) Patulin in Apples and Apple-Based Food Products: The Burdens and the Mitigation Strategies. Toxins (Basel) 10 

14 Brown, J. (2021) The Four Phases of Detox Explained. MosaicDX, Mosaic Diagnostics 

15 Paris, L. (2024) A Functional Medicine Guide to Detox Phases 1, 2, and 3. Paris Healing Arts, Laura Paris 

16 Lynch, T. and Price, A. (2007) The Effect of Cytochrome P450 Metabolism on Drug Response, Interactions, and Adverse Effects. AFP 76, 391–396

17 Li, P., Su, R., Yin, R., Lai, D., Wang, M., Liu, Y., et al. (2020) Detoxification of Mycotoxins through Biotransformation. Toxins (Basel) 12 

18 Ragoubi, C., Quintieri, L., Greco, D., Mehrez, A., Maatouk, I., D’Ascanio, V., et al. (2021) Mycotoxin Removal by spp. and Their Application in Animal Liquid Feed. Toxins (Basel) 13 

19 Ghazvini, R. D., Kouhsari, E., Zibafar, E., Hashemi, S. J., Amini, A. and Niknejad, F. (2016) Antifungal Activity and Aflatoxin Degradation of and Against Toxigenic. Open Microbiol J 10, 197–201 

20 El Adlouni, C., Pinelli, E., Azémar, B., Zaoui, D., Beaune, P. and Pfohl-Leszkowicz, A. (2000) Phenobarbital increases DNA Adduct and Metabolites Formed by Ochratoxin A: Role of CYP 2C9 and Microsomal Glutathione-S-Transferase. Environ Mol Mutagen 35, 123–131 

21 Cook, I., Wang, T. and Leyh, T. S. (2015) Sulfotransferase 1A1 Substrate Selectivity: A Molecular Clamp Mechanism. Biochemistry 54, 6114–6122 

22 Berthiller, F., Crews, C., Dall’Asta, C., Saeger, S. D., Haesaert, G., Karlovsky, P., et al. (2013) Masked Mycotoxins: A Review. Mol Nutr Food Res 57, 165–186 

23 Albillos, A., de Gottardi, A. and Rescigno, M. (2020) The Gut-liver Axis in Liver Disease: Pathophysiological Basis for Therapy. J Hepatol 72, 558–577 

24 Sodani, K., Patel, A., Kathawala, R. J. and Chen, Z.-S. (2012) Multidrug Resistance Associated Proteins in Multidrug Resistance. Chin J Cancer 31, 58–72 

25 Nigam, S. K., Bush, K. T., Martovetsky, G., Ahn, S.-Y., Liu, H. C., Richard, E., et al. (2015) The Organic Anion Transporter (OAT) Family: A Systems Biology Perspective. Physiol Rev 95, 83–123 

26 Dhakal, A. and Sbar, E. (2023) Jarisch-Herxheimer Reaction. In StatPearls, StatPearls Publishing

27 Saad-Hussein, A., Moubarz, G., Mohgah, S. A., Wafaa, G. S. and Aya, H. M. (2019) Role of Antioxidant Supplementation in Oxidant/antioxidant Status and Hepatotoxic Effects due to Aflatoxin B1 in Wheat Miller Workers. J Complement Integr Med 16 

28 Mast Cell Activation Syndrome. Cleveland Clinic 

29 Kritas, S. K., Gallenga, C. E., D Ovidio, C., Ronconi, G., Caraffa, A., Toniato, E., et al. (2018) Impact of Mold on Mast Cell-cytokine Immune Response. J Biol Regul Homeost Agents 32, 763–768

30 Finn, D. F. and Walsh, J. J. (2013) Twenty-first Century Mast Cell Stabilizers. Br. J. Pharmacol. 170, 23–37 

31 (2008) Suppression of Histamine Signaling by Probiotic Lac-B: a Possible Mechanism of Its Anti-allergic Effect. Journal of Pharmacological Sciences, Elsevier 107, 159–166 

Previous post

Leave a comment

Please note, comments must be approved before they are published