Your host and biohacker, Chloe Porter, has a background in engineering, innovation, and research. Her analytical background coupled with her journey in overcoming a brain tumor and defeating several chronic illnesses enables her to approach health and wellness in an innovative way, and now more than ever, she is ready to share her biohacking secrets and expose cutting-edge research. Support this podcast: https://podcasters.spotify.com/pod/show/chloe-porter6/support
In this encore episode, we detail the gut-immune axis, focusing on the protective mucus layer and the role of lactoferrin in intestinal health. We discuss endogenous secretions and how lactoferrin functions as an antimicrobial, immunomodulatory, and iron-regulating glycoprotein, influencing microbial balance and intestinal barrier integrity. Additionally, we highlight lactoferrin testing and levels. We finish by going through supplementation as a phenomenal tool.Topics: 1. The Intestinal Barrier & Mucus Layer- The intestinal lumen, microbiome, mucus layer, epithelial cells, and lamina propria.- The mucus layer: antimicrobial peptides (AMPs) and secretory IgA (sIgA).- Intestinal epithelial cells including goblet cells and enterocytes.2. Immune Cells in the Gut- The lamina propria.- Macrophages, dendritic cells, neutrophils, B cells, T cells, mast cells, and more.3. Antimicrobial Peptides & Secretory IgA- AMPs and sIgA in the mucus layer regulate microbial balance and prevent pathogen adhesion.4. Lactoferrin: Functions & Sources- Iron-binding glycoprotein, antimicrobial and immunomodulatory properties.- Found in mucosal secretions, colostrum, and more.- Intestinal epithelial cells (IECs) and neutrophils.5. Lactoferrin's Role in Intestinal Health- Regulates iron absorption via lactoferrin receptors (LFR)on enterocytes.- Sequesters free iron, limiting bacterial growth, bacteriostatic.- Destabilizes bacterial membranes, cell lysis, bactericidal.6. Lactoferrin Levels- Neutrophils release lactoferrin during infection,increasing its presence in the large intestine.- Fecal lactoferrin levels.- Fecal calprotectin levels.7. Lactoferrin & Candida Overgrowth- Fungal membranes.- Aiding in the inhibition of biofilm formation.8. Lactoferrin Supplementation & Benefits- Supports gut microbiota, epithelial integrity, and immune modulation.- Bovine colostrum and as a supplement.9. Conclusion- The intestinal mucus layer is key to gut barrier function.- Lactoferrin plays a central role in microbial regulation and immune defense.Get Chloe's Book Today! "75 Gut-Healing Strategies & Biohacks" Follow Chloe on Instagram @synthesisofwellnessVisit synthesisofwellness.com
In this encore episode, we go through the pathophysiology of hydrogen sulfide (H₂S)-dominant small intestinal bacterial overgrowth (SIBO), focusing on its role in intestinal motility disruption, microbial dysbiosis, and epithelial barrier dysfunction. We detail the interplay between the enteric nervous system (ENS), interstitial cells of Cajal (ICCs), and migrating motor complex (MMC) in regulating small intestinal transit and how excess H₂S modulates smooth muscle activity, inflammatory signaling, and gastrointestinal transit time. Lastly, we detail symptoms and related SIBO subtypes.Topics:1. Introduction to Hydrogen Sulfide Dominant SIBO - Overview of hydrogen sulfide (H₂S) SIBO - Sulfate as an electron acceptor - H₂S production, sulfur-containing amino acids - Physiological roles of H₂S vs. in excess 2. Hydrogen Sulfide and Gastrointestinal Motility - Impact on transit time - Association with IBS-like symptoms, diarrhea 3. Intestinal Motility and Regulation - Role of the ENS, ICCs, smooth muscle cells, and neurotransmitters in motility 4. The Enteric Nervous System (ENS) and Gut Motility - ENS as the "second brain" and its control over digestion - Myenteric and submucosal plexuses: regulation of peristalsis, secretion, blood flow - Gut-brain axis involvement via vagus nerve signaling - ICCs as pacemaker cells generating slow-wave electrical activity - MMC's role in clearing debris and bacteria 5. Dysregulated Motility and SIBO Development 6. Overlapping Symptoms Across SIBO Subtypes - Shared symptoms - Hydrogen and H₂S-dominant SIBO vs. IMO 7. Gastric Acid and Its Role - HCl secretion by parietal cells and its role in digestion and microbial defense - Hypochlorhydria and bacterial overgrowth 8. Conclusion - Root cause approach.Get Chloe's Book Today! "75 Gut-Healing Strategies & Biohacks" Follow Chloe on Instagram @synthesisofwellnessVisit synthesisofwellness.com
In this episode, we detail the oral microbiome and its relationship with the intestinal microbiome, detailing the oral-gut microbiota axis. We discuss the stepwise development of oral biofilm, the transition from plaque to dental calculus, and potential systemic impacts of oral dysbiosis, while highlighting intestinal health and focusing on Fusobacterium nucleatum. We then detail possible symptoms of oral dysbiosis as well as testing modalities. Topics:1. Overview of the Oral Microbiome and the Oral-Gut Axis - The oral cavity and the gut: microbially dense sites. - These regions are anatomically distinct but connected and capable of influencing each other. - The oral microbiome: bacterial species, fungi, viruses, archaea, protozoa. - Microbial distribution is influenced by the topography and chemistry of various oral surfaces. 2. Oral Cavity Anatomy and Microenvironments - The oral cavity is divided into the vestibule and oral cavity proper. - Lined by oral mucosa, which varies in structure. - These anatomical variations create unique ecological niches that support different microbial populations. 3. Tooth Structure and Relevance to Microbial Colonization - Teeth are embedded in the alveolar processes of the maxilla and mandible. - Tooth structure. - Long-term microbial colonization and biofilm development. 4. Biofilm / Dental Plaque Development - Biofilm begins with the formation of the acquired pellicle.- Pioneer species secreting extracellular polymeric substances (EPS).- Secondary colonizers coaggregate.- Coaggregation is species-specific.- Example.- Aerobes can lower local oxygen levels, creating micro anaerobic niches that support obligate anaerobes. 5. Maturation of the Biofilm - Biofilm composition shifting to anaerobic, proteolytic species such as P. gingivalis. - F. nucleatum: a bridge species. 6. Mineralization and Formation of Dental Calculus - Mineralization, calcium and phosphate deposition. - Dental calculus. - Can promote further accumulation and worsen inflammation. 7. Oral Dysbiosis - An imbalance in the oral microbial community. - Potential signs and symptoms. - Halitosis, swollen or bleeding gums, tooth sensitivity, new cavities, dry mouth or altered taste, recurrent infections, coated tongue, plaque buildup near the gumline. 8. Oral-Gut Axis and Systemic Implications - Physical, chemical, biochemical, and immunological barriers aid in preventing oral bacteria from colonizing the gut. - Oral bacteria can potentially impact intestinal health. - F. nucleatum and IBD. 9. Oral Microbiome Testing10. Multi-Factorial Get Chloe's Book Today! "75 Gut-Healing Strategies & Biohacks" Follow Chloe on Instagram @synthesisofwellnessVisit synthesisofwellness.com
In this encore episode, we detail the intestinal mycobiome and the mechanisms by which beneficial microbes aid in regulating opportunistic fungal species: Candida albicans. We discuss Candida overgrowth virulence and how disruptions in microbial balance, immune function, and epithelial integrity impact Candida's pathogenicity. Additionally, we discuss the role of mast cells in the lamina propria, their involvement in mucosal immunity, and how Mast Cell Activation Syndrome (MCAS) can be exacerbated by fungal dysbiosis / microbial imbalances.Topics: 1. Introduction - The intestinal mycobiome and its role in intestinal health. - Regulating populations of opportunistic fungal species like Candida albicans.- Mast cell activation syndrome (MCAS) intro. 2. Structure of the Intestinal Barrier- The microbiome and mucus layer: microbes, mucins, antimicrobial peptides, sIgA.- The epithelial monolayer: enterocytes, goblet cells, Paneth cells, enteroendocrine cells…- Tight junction proteins and paracellular transport.- The lamina propria beneath the epithelium: immune cells and more. 3. Candida's Pathogenicity and Barrier Disruption- Exists primarily in its yeast form, regulated by microbial competition, host immune defenses, antifungal peptides. - Secretory IgA (sIgA) and immune surveillance mechanisms help regulate fungal populations. - Chronic stress, immune suppression, and dysbiosis can deplete sIgA, increasing susceptibility to Candida proliferation. - Environmental triggers such as immune suppression, microbial depletion, inflammatory cytokines.- Yeast-to-hyphal transition, deeper tissue invasion. - Secreted aspartyl proteinases (SAPs), phospholipases, candidalysin: epithelial damage.- Pattern recognition receptors (PRRs) and overgrowth detection. 4. Impact of Beneficial Bacteria on Candida Overgrowth - Beneficial bacteria compete with Candida for nutrients and epithelial adhesion sites. - Short-chain fatty acids (SCFAs) produced by beneficial bacteria.- Depletion of beneficial bacteria removes ecological resistance, allowing Candida to proliferate unchecked. 5. Mast Cells in the Lamina Propria and Their Role in Intestinal Immunity- Mast cell location - Upon activation, mast cells release histamine, cytokines, proteases, and more that regulate gut immune responses. - Histamine can increase gut permeability and modulate local immune activation. - Tryptase and chymase. - Pro-inflammatory cytokines. 6. Mast Cell Activation Syndrome (MCAS) - Mast cell hyperactivation.- Environmental triggers, toxins, chronic infections, and stress. - Candida overgrowth and mast cell activation.- Addressing microbial imbalances and reducing the body's total microbial, chemical, and toxin burden.- A diverse microbiome. 7. Conclusion- Candida overgrowth and regulation via immune defenses and beneficial bacterial competition.- MCAS, root cause approach.Get Chloe's Book Today! "75 Gut-Healing Strategies & Biohacks" Follow Chloe on Instagram @synthesisofwellnessVisit synthesisofwellness.com
In this encore episode, we detail the pathophysiology of intestinal methanogen overgrowth (IMO), hydrogen-dominant small intestinal bacterial overgrowth (H₂-SIBO), and hydrogen sulfide-dominant SIBO (H₂S-SIBO), discussing their mechanistic interactions and overlap. We discuss methanogenic archaea, as well hydrogen sulfide- and hydrogen-producing microbes and their contributions to altered gastrointestinal motility, epithelial barrier function, and neuromuscular signaling. We detail host endogenous defense mechanisms, including gastric acid secretion, pancreatic enzymatic activity, bile flow, intestinal motility, and more.Topics:1. Introduction- Overview of intestinal methanogen overgrowth (IMO), hydrogen sulfide-dominant SIBO, and hydrogen-dominant SIBO - Discussion of overlap 2. Intestinal Methanogen Overgrowth (IMO)- Characterized by an overabundance of methane-producing archaea - Methane and intestinal transit- Bloating, abdominal discomfort, constipation - Associated with irritable bowel syndrome with constipation (IBS-C) 3. Overlap Between IMO and SIBO Subtypes- Hydrogen as a substrate for methane and/or hydrogen sulfide production- Shared potential root causes 4. Small Intestinal Bacterial Overgrowth (SIBO) and Protective Mechanisms- The small intestine remains relatively free of bacteria due to protective mechanisms - Gastric acid secretion - Pancreatic enzymes and bile - Intestinal motility- Structural abnormalities 5. Hydrogen-Dominant SIBO - Increased hydrogen and intestinal transit- Potential symptoms, diarrhea, postprandial bloating - Different forms of SIBO can coexist 6. Hydrogen Sulfide-Dominant SIBO - H₂S and intestinal motility- H₂S and intestinal epithelial integrity- Symptoms, associations with IBS diarrhea 7. Host Defense Mechanisms Regulating Microbial Balance- Gastric acid secretion, pancreatic enzyme activity, bile flow, intestinal motility, and more 8. Gastric Acid and Its Role in Microbial Regulation- Parietal cells secrete hydrochloric acid (HCl) and intrinsic factor - Chief cells secrete pepsinogen, conversion into pepsin in acidic conditions - Gastric acid, digestion and antimicrobial defense 9. Hypochlorhydria - Low gastric acid impairs microbial defense- Reduced acidity disrupts digestion and downstream pancreatic enzyme and bile release 10. Pancreatic Enzymes and Their Role in Microbial Regulation- Pancreatic enzymes, digestion, antimicrobial activity - Enzymes modify chyme to limit fermentable substrates that fuel microbial proliferation 11. Intestinal Motility - Coordinated contractions propel food, microbes, and waste through the GI tract - The enteric nervous system (ENS) and gut motility- The myenteric plexus controls peristalsis, while the submucosal plexus regulates secretion and absorption 12. Conclusion- Intestinal methanogen overgrowth (IMO), hydrogen-dominant SIBO, and hydrogen sulfide-dominant SIBO - Overlap in symptoms and microbial interactions - Host defense mechanisms, including gastric acid, pancreatic enzymes, and motility - Intestinal motility and ENS function in microbial homeostasis - Hydrogen sulfide as a gasotransmitterGet Chloe's Book Today! "75 Gut-Healing Strategies & Biohacks" Follow Chloe on Instagram @synthesisofwellnessVisit synthesisofwellness.com
In this episode, we highlight the stomach's role in vitamin B12 absorption and bioavailability, detailing the cellular composition of the gastric mucosa and further highlighting mucous cells, parietal cells, and chief cells. We also briefly discuss Helicobacter pylori and common symptoms, while detailing H. pylori's survival mechanisms in the acidic gastric environment as well as potential impacts on the gastric mucosal barrier. Topics:1. Introduction- Overview of the stomach's role in B12 absorption- Helicobacter pylori 2. H. pylori Overview- Gram-negative bacterium, can colonize the stomach lining- Many individuals remain asymptomatic- Symptoms - Can contribute to gastritis and peptic ulcers- Produces urease, hydrolyzes urea into ammonia (NH₃) andcarbon dioxide (CO₂)- Ammonia neutralizes stomach acid locally, protective microenvironment- H. pylori damages the mucosal barrier and contributes to persistence- Possesses additional virulence factors 3. Gastric Anatomy - Stomach is divided into the cardia, fundus, body, and pylorus- The gastric mucosa - The epithelial lining; mucous cells, parietal cells, chief cells, and more 4. Mucous Cells and Mucosal Protection - Line the gastric pits and secrete thick, viscous mucus- Mucus composed of water, mucin glycoproteins, and other low-molecular-weight molecules- Traps bicarbonate ions (HCO₃⁻)- Shields the stomach lining 5. Parietal Cells - Located in the gastric glands, predominantly in the fundus and body- Secrete hydrochloric acid (HCl)- Secrete intrinsic factor (IF) 6. Vitamin B12 Overview - Cobalamin: DNA synthesis, red blood cell formation, neurological function, methylation, and more- Methylcobalamin and 5-deoxyadenosylcobalamin- Non-active forms include hydroxocobalamin and synthetic cyanocobalamin 7. Vitamin B12 Absorption Pathway - B12 is released from proteins by HCl and pepsin- Binds first to haptocorrin- In the small intestine, pancreatic enzymes degrade haptocorrin- B12 binds intrinsic factor - Absorption in the ileum - Impairment 8. Chief Cells - Located in the gastric glands- Secrete pepsinogen, activated by acid into pepsin- Also secrete gastric lipase 9. Hydrochloric Acid (HCl) - Secreted by parietal cells- Denatures dietary proteins and activates pepsinogen- Acts as a defense mechanism by aiding in sterilizing ingested food 10. Relevance of H. pylori - Produces urease that breaks down urea into NH₃ and CO₂- Ammonia neutralizes acid locally, forming a protective “bubble”- Enhances mucosal damage and microbial persistence 11. B12 Absorption - Multiple factors and root causes can impair absorption- H. pylori 12. Hypochlorhydria - Symptoms- Absorption: vitamin B12, iron, calcium, magnesium, more- Small Intestinal Bacterial Overgrowth (SIBO) 13. Conclusion- Multi-factorial, root cause approachThanks for tuning in!Get Chloe's Book Today! "75 Gut-Healing Strategies & Biohacks" Follow Chloe on Instagram @synthesisofwellnessFollow Chloe on TikTok @chloe_c_porterVisit synthesisofwellness.com to purchase products, subscribe to our mailing list, and more!
In this episode, we highlight vitamin D₃ in gastrointestinal health, with a focus on intestinal barrier function and the gut-immune axis. We discuss how vitamin D is metabolized into its active form, 1,25-dihydroxyvitamin D, and how 1,25-dihydroxyvitamin D can support the intestinal mucosa, while highlighting the intestinal epithelium and immune cells in the lamina propria. Through this, we further detail the role of regulatory T cells in supporting immune balance and modulating immune function.Topics:1. Introduction - Highlighting intestinal barrier function and the gut-immune axis with a focus on vitamin D₃ 2. Vitamin D Synthesis and Metabolism - Skin and 7-dehydrocholesterol; dietary intake.- Transported through the bloodstream bound to vitamin D binding protein.- First hydroxylation in the liver to form 25-hydroxyvitamin D₃ (25(OH)D₃).- Testing.- Second hydroxylation in the kidney to form the active 1,25 dihydroxyvitamin D₃ (1,25(OH)₂D₃).- Local activation also occurs. 3. Intestinal Barrier Anatomy - The intestinal epithelium forms a selectively permeable monolayer that interfaces with the gut lumen.- Covered by a mucus layer: mucins, antimicrobial peptides, secretory IgA.- Mucus layer provides physical and biochemical protection.- Tight junction proteins and paracellular permeability.- Molecules crossing the epithelium. - Disruption of tight junctions, immune cell exposure to luminal contents.- The lamina propria and Gut-Associated Lymphoid Tissue (GALT). - Maintaining tolerance while defending against pathogens. 4. Vitamin D's Role in Barrier Support and Immune Modulation - 1,25-dihydroxyvitamin D₃, tight junction integrity, immune regulation, and more.- Regulatory T Cells (Tregs) and immune balance. 5. Vitamin D₂ and D₃- D₃ (cholecalciferol) is more biologically effective and binds DBP more strongly. 6. Vitamin D Levels - Levels observed in autoimmune conditions, IBS, and more.- Symptoms.- Insufficient and excessive levels of vitamin D can be detrimental.- Multi-system. 7. Conclusion - Multifactorial: Numerous factors regulate vitamin D levels.- Gastrointestinal health root cause approach.Thanks for tuning in!Get Chloe's Book Today! "75 Gut-Healing Strategies & Biohacks" Follow Chloe on Instagram @synthesisofwellnessFollow Chloe on TikTok @chloe_c_porterVisit synthesisofwellness.com to purchase products, subscribe to our mailing list, and more!
In this episode, we discuss the gut-immune axis and Small Intestinal Fungal Overgrowth (SIFO), with a mechanistic focus on immune activation and epithelial barrier disruption. We detail contributing factors to and symptoms of SIFO. We further discuss the anatomical positioning and immunological functions of mast cells within the lamina propria, highlighting mast cell activation syndrome, while detailing how SIFO and SIBO can serve as upstream triggers for mast cell activation.Topics:1. The Gut-Immune Axis and Mast Cells - The intestinal epithelium forms the innermost selective barrier, coated in a protective mucus layer.- Immediately beneath lies the lamina propria, rich in immune cells including mast cells. 2. Mast Cells in the Lamina Propria - Mast cells are positioned near nerves, capillaries, lymphatics, and epithelial cells within the lamina propria.- Their anatomical location enables rapid immune surveillance and inflammatory response.- Mast cell granules store histamine and more. 3. Mast Cell Mediators - Histamine: Biogenic amine, inflammatory signaling.- Tryptase: A serine protease.- Cytokines and lipid mediators also released upon activation. 4. Mast Cell Activation Syndrome (MCAS) - Systemic: skin, respiratory tract, cardiovascular system, nervous system, and more.- The gastrointestinal tract.- Common triggers for mast cell activation. 5. Transition to Small Intestinal Fungal Overgrowth (SIFO) - Overgrowth of fungal organisms, often candida species, within the small intestine.- Contributing factors including hypochlorhydria, slowed motility, immune suppression, disrupted bacterial populations.- Overlap: SIFO, SIBO 6. Hypochlorhydria and the Stomach's Defense Role - Parietal cells secrete hydrochloric acid (HCl) and intrinsic factor.- HCl denatures proteins and sterilizes ingested pathogens.- Chief cells secrete pepsinogen, which becomes pepsin in acidic conditions to digest proteins.- Consequences of Reduced Gastric Acidity 7. The Role of Intestinal Motility - Enteric nervous system (ENS), myenteric and submucosal plexuses.- Coordinated contractions preventing stasis and microbial overgrowth. 8. Common Symptoms of SIFO and clinical overlap with SIBO 9. Candida and Fungal Pathophysiology in the Gut - Yeast form, regulated by microbial competition and immune defenses.- Hyphal transformation.- Degrade the mucus layer, disrupt epithelial integrity, trigger inflammation. 10. Secretory IgA and Mucosal Defense - Chronic stress, immune dysfunction, dysbiosis can lower sIgA levels and weaken mucosal immunity. 11. SIFO, SIBO, and Mast Cell Activation 12. Conclusion - Root cause approach, multi-factorial Thanks for tuning in!Get Chloe's Book Today! "75 Gut-Healing Strategies & Biohacks" Follow Chloe on Instagram @synthesisofwellnessFollow Chloe on TikTok @chloe_c_porterVisit synthesisofwellness.com to purchase products, subscribe to our mailing list, and more!
In this episode, we detail the role of vitamin B1 (thiamine) in supporting gastrointestinal function, highlighting intestinal motility through vitamin B1's involvement in acetyl-CoA and acetylcholine synthesis. We discuss thiamine insufficiency as well as how small intestinal bacterial overgrowth (SIBO) can further affect thiamine availability. We further detail host defense mechanisms—including gastric acid, pancreatic enzymes, and gastrointestinal motility—that work in concert to regulate microbial balance in the small intestine.Topics: 1. Introduction- Focus on SIBO, intestinal motility, and vitamin B1 (thiamine) 2. Small Intestine and Microbial Regulation- Low bacterial load under healthy conditions - Protective mechanisms: gastric acid, enzymes, motility… 3. Overview of SIBO Subtypes - H₂-SIBO, hydrogen-producing bacteria - IMO, methane-producing archaea - H₂S-SIBO, hydrogen sulfide-producing bacteria - Subtypes can coexist, share underlying factors 4. Host Factors in Microbial Regulation - Gastric acid, motility, pancreatic enzymes, bile acids, and immune surveillance 5. Gastric Acid and Parietal Cells - Hydrochloric acid and intrinsic factor - Acid environment supports digestion and microbial regulation - Protein denaturation and pepsin activation 6. Chief Cells and Enzyme Function - Pepsinogen and gastric lipase - Pepsinogen activated by acidic pH 7. Hypochlorhydria and Downstream Effects 8. Pancreatic Enzymes in the Small Intestine 9. Intestinal Motility and the Enteric Nervous System (ENS)- Coordinated smooth muscle contractions - Myenteric and submucosal plexuses 10. Intestinal Wall Anatomy - Epithelium and lamina propria - Submucosa contains the submucosal plexus - Myenteric plexus located between muscle layers 11. Vitamin B1 (Thiamine) and Gastrointestinal Function - Acetylcholine synthesized from choline and acetyl-CoA - Acetyl-CoA formation, thiamine availability - Acetylcholine signaling 12. SIBO and Nutrient Availability- Bacterial overgrowth can affect nutrient absorption - Thiaminases 13. Conclusion - Root cause approach, multi-factorial Thanks for tuning in!Get Chloe's Book Today! "75 Gut-Healing Strategies & Biohacks" Follow Chloe on Instagram @synthesisofwellnessFollow Chloe on TikTok @chloe_c_porterVisit synthesisofwellness.com to purchase products, subscribe to our mailing list, and more!
In this encore episode, we discuss the role of histamine within the gastrointestinal tract, detailing its regulation of gastric acid secretion and impact on intestinal motility. We examine how histamine-producing enterochromaffin-like (ECL) cells, mast cells, and gut microbiota contribute to histaminergic signaling, and how enzymatic degradation via diamine oxidase (DAO) maintains homeostasis. Additionally, we discuss histamine intolerance, highlighting the impacts of dysbiosis (as well as SIBO or small intestinal bacterial overgrowth) and intestinal barrier dysfunction on DAO insufficiency and histamine accumulation.Topics: 1. Introduction to Histamine & the Gastrointestinal Tract- Role in digestion and motility- Impact of intestinal conditions such as SIBO 2. Histamine as a Biogenic Amine- Definition and classification- L-histidine and histidine decarboxylase (HDC)- Presence in various tissues, including the GI tract 3. Sources of Histamine in the Gut- Enterochromaffin-like (ECL) cells in the stomach- Mast cells in the lamina propria- Histamine-producing gut bacteria 4. Histamine's Role in Digestion & Gastric Acid Secretion- Interaction with parietal cells in the stomach- Activation of proton pumps via H₂ receptor binding- Maintenance of acidic pH for digestion- Importance of proper pH within the stomach for protecting against pathogenic microbes- Impact on downstream gut microbiota balance (e.g., SIBO) 5. Histamine & Intestinal Motility- Modulation of gut motility via H₁ and H₃ receptors- H₁ receptor activation leading to contractions (diarrhea, cramping)- H₃ receptor-mediated inhibition of neurotransmitter release (bloating, slow motility) 6. Histamine Intolerance: Definition & Mechanisms- Diamine Oxidase and histamine breakdown- DAO secretion by enterocytes (intestinal epithelial cells)- Enzymatic breakdown of histamine before absorption 7. Factors That Can Affect DAO Activity- Genetic Polymorphisms: AOC1 gene, reduced DAO expression- Cofactor Deficiencies: Copper, vitamin B6, and vitamin C as essential DAO cofactors- Importance of intestinal lining integrity for DAO production- Conditions leading to or associated with enterocyte damage- Intestinal dysbiosis and histamine accumulation- Intestinal dysbiosis & epithelial/enterocyte damage- Suppression of DAO production due to epithelial dysfunction- Small Intestinal Bacterial Overgrowth (SIBO) & Histamine 8. Symptoms of Histamine Intolerance- Gastrointestinal symptoms (bloating, gas, constipation, diarrhea, acid reflux...)- Systemic symptoms (dizziness, flushing, migraines...) 9. Identifying Potential Root Causes- Tools and supplements- Bioindividual approachThanks for tuning in!Get Chloe's Book Today! "75 Gut-Healing Strategies & Biohacks" Follow Chloe on Instagram @synthesisofwellnessFollow Chloe on TikTok @chloe_c_porterVisit synthesisofwellness.com to purchase products, subscribe to our mailing list, and more!
In today's episode, we detail the enteric nervous system and regulation of gastrointestinal motility. We discuss factors including dysautonomia, stress, microbial overgrowth, and more, with regards to potential effects upon gastrointestinal motility. We further detail symptoms of altered GI motility. Topics:1. Introduction to Gastrointestinal Motility- Orchestrated contraction of smooth muscles that propel contents along the digestive tract. - Roles in mixing, absorption, and preventing bacterial overgrowth.- Disruptions in motility. 2. The Enteric Nervous System (ENS) and Its Role- Myenteric and submucosal plexus.- Coordinating contractions and relaxations for effective motility. - Mucosa includes epithelium, lamina propria, and muscle. - Submucosa houses the submucosal plexus. - Muscularis externa. - Inner circular and outer longitudinal muscle layers with the myenteric plexus in between. 3. The Role of Interstitial Cells of Cajal (ICCs)- Specialized pacemaker cells in muscular layers - Generate slow-wave electrical activity to synchronize smooth muscle contractions - Critical for peristalsis.- Work with neural inputs to fine-tune gut motility 4. Dysautonomia and Its Impact on Gut Motility- Dysfunction of the autonomic nervous system (ANS) - Motility impacts- Microbial overgrowth, SIBO 5. Gastrointestinal Dysmotility- Neurological dysregulation - Structural abnormalities and smooth muscle dysfunction - Hormonal imbalances - Microbial overgrowth - Autoimmunity 6. The HPA Axis- Hypothalamus, pituitary gland, adrenal glands - Central to the stress response and interlinked with gut function - The amygdala and PVN of the hypothalamus - CRH stimulates ACTH, leading to cortisol release from adrenal glands 7. Cortisol and Gut Barrier Function- Cortisol is a glucocorticoid that modulates immune function and gut physiology - Chronic cortisol exposure can impair tight junction (TJ) integrity - Increases paracellular permeability and allows passage of antigens and endotoxins 8.Stress-Induced Changes in Gut Motility- Slow gastric motility - Increased colonic motility 9. Symptoms of GI Dysmotility10. Addressing Root Cause(s)Thank you to our episode sponsor:1. Shop Fresh Press Farms'Peach Cider Vinegar at Sprouts locations nationwide, and check out their full collection here. Get Chloe's Book Today! "75 Gut-Healing Strategies & Biohacks" Follow Chloe on Instagram @synthesisofwellnessVisit synthesisofwellness.com
In this episode, we detail the cellular make-up of the gastric lining, highlighting hydrochloric acid (HCl) secretion and its role in digestion, micronutrient absorption, and microbial defense. We discuss potential pathophysiological consequences of hypochlorhydria, including impaired digestive signaling, bacterial overgrowth in the small intestine (SIBO), and more. Lastly, we detail upstream microbial control, focusing on the influence of the oral microbiome and oral dysbiosis.Topics: 1. Introduction- Highlighting the stomach and hypochlorhydria / low stomach acid, as well as microbial balance, dysbiosis, and overgrowth.- Discussion of intestinal microbiome and oral microbiome. 2. Gastric Anatomy and Mucosal Layers- The stomach: cardia, fundus, body, pylorus.- The gastric mucosa: epithelial lining, lamina propria, muscularis mucosae.- The epithelial layer: mucous cells, parietal cells, chief cells, enteroendocrine cells.- Beneath the mucosa: submucosa and muscular layers that contribute to gastric motility, mechanical digestion, and more. 3. Mucous Cells - The surface epithelium and gastric pits.- Secretion of thick, viscous mucus.- Protecting the epithelium.- The mucus traps bicarbonate ions: neutral microenvironment that protects against acid-induced damage. 4. Parietal Cells and Gastric Acid Secretion- Secretion of hydrochloric acid (HCl): denatures dietary proteins, sterilizes ingested food.- Parietal cells produce intrinsic factor: binds vitamin B12 to facilitate absorption in the ileum. 5. Chief Cells and Pepsinogen- Secretion of pepsinogen: activated by low pH into the proteolytic enzyme pepsin.- Pepsin and protein digestion.- Release of gastric lipase. 6. Hydrochloric Acid: Digestive and Protective Roles- Immune defense.- Pepsinogen activation.- Adequate gastric acidity essential for properly acidified chyme.- Priming release of bile and pancreatic enzymes. 7. The Small Intestine, Stomach, and Microbial Regulation- The pyloric sphincter.- The stomach supports minimal microbial diversity.- Acidic barrier aids in limiting the entry of opportunistic microbes into the small intestine. 8. Hypochlorhydria, Low Stomach Acid- Hypochlorhydria can compromise one of the body's defense mechanisms.- Colonization of the small intestine.- Insufficient pepsinogen activation and possible protein digestion impairment.- Suboptimal chyme formation and downstream digestive capacity.- Possible symptoms of hypochlorhydria.- Possible impaired absorption of micronutrients including vitamin B12, iron, calcium, magnesium.- H. pylori infection, autoimmunity, aging. 9. The Oral Microbiome, Intestinal Microbiome, and Dysbiosis- Oral and environmental microbes.- Altered gastric pH, bile, digestive enzymes, and more.- The oral cavity as the gateway to the GI tract.- Oral microbial species.- Oral hygiene and oral microbiome health. 10. Conclusion- Root-cause approach.Thank you to our episode sponsor:1. Shop Fresh Press Farms'Peach Cider Vinegar at Sprouts locations nationwide, and check out their full collection here. Get Chloe's Book Today! "75 Gut-Healing Strategies & Biohacks" Follow Chloe on Instagram @synthesisofwellnessVisit synthesisofwellness.com
We are honored to be joined by Forrest Smith, the CEO and Founder of Kineon. Forrest has a 20-year history of building successful startups in tech hardware. He is passionate about health, wellness, and advancing technology to help others, and he has spent his adult life building remarkable products around innovative supply chain. Aside from his founder position in multiple companies, Forrest has held several positions, from Director of Asian Operations for a Fortune 500 company to Director of Business Development. He also grew up playing competitive sports in Atlanta, regularly participates in rugby matches, and trains CrossFit, which ultimately led him to develop Kineon Lab's Move+, a modular, targeted laser therapy device for neuromuscular pain and inflammation.Topics:1. Forrest's Personal Experience with Red Light Therapy - The creation of Kineon and the MOVE+ Pro. 2. Cellular Health - Supporting healthy mitochondrial function. - ATP production and oxidative stress.- The role of oxidative stress in impairing mitochondrial output. - Balancing ROS production and cellular repair. 3. Red and Infrared Light Therapy- Photoacceptors including cytochrome c oxidase. - Nitric oxide.- Comparing wavelengths and their tissue penetration. - Clinical research on red light therapy.- Joint health, mobility, and blood flow.- Skin and hair scientific literature. - Technical specifications. - Importance of dose control.- LEDs vs. Lasers in light therapy devices. - Including laser technology in the MOVE+ Pro. 4. Kineon Lab's MOVE+ Pro - Remarkable engineering and performance.- Modular design for targeting joints and soft tissue. - Joint pain, inflammation, and recovery. - How often and how long to use the MOVE+ Pro. - Sample protocols for joint pain and performance recovery. Shop the Kineon Move+ Pro and save $100 during Kineon's Spring Amazon Sale running from March 25th to March 31stCheck out Kineon's website hereCheck out Kineon's Instagram @kineon_labsGet Chloe's Book Today! "75 Gut-Healing Strategies & Biohacks" Follow Chloe on Instagram @synthesisofwellnessVisit synthesisofwellness.com
In this episode, we detail the intestinal mycobiome and the mechanisms by which beneficial microbes aid in regulating opportunistic fungal species: Candida albicans. We discuss Candida overgrowth virulence and how disruptions in microbial balance, immune function, and epithelial integrity impact Candida's pathogenicity. Additionally, we discuss the role of mast cells in the lamina propria, their involvement in mucosal immunity, and how Mast Cell Activation Syndrome (MCAS) can be exacerbated by fungal dysbiosis / microbial imbalances.Topics: 1. Introduction - The intestinal mycobiome and its role in intestinal health. - Regulating populations of opportunistic fungal species like Candida albicans.- Mast cell activation syndrome (MCAS) intro. 2. Structure of the Intestinal Barrier- The microbiome and mucus layer: microbes, mucins, antimicrobial peptides, sIgA.- The epithelial monolayer: enterocytes, goblet cells, Paneth cells, enteroendocrine cells…- Tight junction proteins and paracellular transport.- The lamina propria beneath the epithelium: immune cells and more. 3. Candida's Pathogenicity and Barrier Disruption- Exists primarily in its yeast form, regulated by microbial competition, host immune defenses, antifungal peptides. - Secretory IgA (sIgA) and immune surveillance mechanisms help regulate fungal populations. - Chronic stress, immune suppression, and dysbiosis can deplete sIgA, increasing susceptibility to Candida proliferation. - Environmental triggers such as immune suppression, microbial depletion, inflammatory cytokines.- Yeast-to-hyphal transition, deeper tissue invasion. - Secreted aspartyl proteinases (SAPs), phospholipases, candidalysin: epithelial damage.- Pattern recognition receptors (PRRs) and overgrowth detection. 4. Impact of Beneficial Bacteria on Candida Overgrowth - Beneficial bacteria compete with Candida for nutrients and epithelial adhesion sites. - Short-chain fatty acids (SCFAs) produced by beneficial bacteria.- Depletion of beneficial bacteria removes ecological resistance, allowing Candida to proliferate unchecked. 5. Mast Cells in the Lamina Propria and Their Role in Intestinal Immunity- Mast cell location - Upon activation, mast cells release histamine, cytokines, proteases, and more that regulate gut immune responses. - Histamine can increase gut permeability and modulate local immune activation. - Tryptase and chymase. - Pro-inflammatory cytokines. 6. Mast Cell Activation Syndrome (MCAS) - Mast cell hyperactivation.- Environmental triggers, toxins, chronic infections, and stress. - Candida overgrowth and mast cell activation.- Addressing microbial imbalances and reducing the body's total microbial, chemical, and toxin burden.- A diverse microbiome. 7. Conclusion- Candida overgrowth and regulation via immune defenses and beneficial bacterial competition.- MCAS, root cause approach.Thank you to our episode sponsor:1. Shop Fresh Press Farms'Peach Cider Vinegar at Sprouts locations nationwide, and check out their full collection here. Get Chloe's Book Today! "75 Gut-Healing Strategies & Biohacks" Follow Chloe on Instagram @synthesisofwellnessVisit synthesisofwellness.com
In this episode, we detail the role of immunoglobulins in gut-immune function, examining their involvement in mucosal defense, microbial regulation, and immune homeostasis within the intestines. We discuss the mechanisms of antibody production, highlighting how secretory IgA (sIgA) contributes to intestinal barrier integrity and pathogen neutralization. We also detail low secretory immunoglobulin A levels and Candida overgrowth. Lastly, we go through tools to support the gut-immune axis including oral immunoglobulin supplementation.1. Introduction - Overview of immunoglobulins, roles in gut-immune health - The gut as a constant interface with antigens, microbes, and potential pathogens - Adequate but balanced immunoglobulin levels; preventing opportunistic pathogen overgrowth 2. Immunoglobulins - Immunoglobulins (Ig) as glycoproteins, fundamental role in immune defense - Five major immunoglobulin isotypes and their distinct roles - How immunoglobulins recognize and neutralize pathogens / toxins / foreign antigens 3. Antibody Production in the Intestines - Naïve B cells originate in the bone marrow, migrate to secondary lymphoid tissues - Antigen-presenting cells (APCs) facilitate antigen processing and B cell activation - Plasma cells and mucosal immunity 4. The Intestinal Barrier and Immune Components - Layers of the gut barrier and their functions - The role of gut-associated lymphoid tissue (GALT) in immune surveillance - Immune cells within the lamina propria and their contributions to gut homeostasis 5. Secretory IgA (sIgA) and Its Role in Gut Immunity - sIgA production and transport across the intestinal epithelium - Role of sIgA in immune exclusion and pathogen neutralization 6. Factors That Can Influence sIgA Levels - Chronic stress, infections, and gut inflammation as contributors to low sIgA - Consequences of low sIgA, including increased susceptibility to pathogens - Nutrients essential for supporting optimal sIgA levels 7. Other Key Immunoglobulins in Gut Immunity - Immunoglobulin G's role in immune responses / pathogen neutralization - Immunoglobulin M's location and function as an early immune responder in mucosal defense 8. Supplemental Immunoglobulins - Oral immunoglobulin supplementation and its benefits - Bovine colostrum as a source of immunoglobulins for intestinal and immune support 9. Conclusion - Recap of immunoglobulins' role in gut-immune function - The importance of balanced IgA, IgG, and IgM levels - Strategies to support gut immunityThank you to our episode sponsor:1. Shop CYLN's full skincare line here.Get Chloe's Book Today! "75 Gut-Healing Strategies & Biohacks" Follow Chloe on Instagram @synthesisofwellnessVisit synthesisofwellness.com
In this episode, we detail the pathophysiology of intestinal methanogen overgrowth (IMO), hydrogen-dominant small intestinal bacterial overgrowth (H₂-SIBO), and hydrogen sulfide-dominant SIBO (H₂S-SIBO), discussing their mechanistic interactions and overlap. We discuss methanogenic archaea, as well hydrogen sulfide- and hydrogen-producing microbes and their contributions to altered gastrointestinal motility, epithelial barrier function, and neuromuscular signaling. We detail host endogenous defense mechanisms, including gastric acid secretion, pancreatic enzymatic activity, bile flow, intestinal motility, and more.Topics:1. Introduction- Overview of intestinal methanogen overgrowth (IMO), hydrogen sulfide-dominant SIBO, and hydrogen-dominant SIBO - Discussion of overlap 2. Intestinal Methanogen Overgrowth (IMO)- Characterized by an overabundance of methane-producing archaea - Methane and intestinal transit- Bloating, abdominal discomfort, constipation - Associated with irritable bowel syndrome with constipation (IBS-C) 3. Overlap Between IMO and SIBO Subtypes- Hydrogen as a substrate for methane and/or hydrogen sulfide production- Shared potential root causes 4. Small Intestinal Bacterial Overgrowth (SIBO) and Protective Mechanisms- The small intestine remains relatively free of bacteria due to protective mechanisms - Gastric acid secretion - Pancreatic enzymes and bile - Intestinal motility- Structural abnormalities 5. Hydrogen-Dominant SIBO - Increased hydrogen and intestinal transit- Potential symptoms, diarrhea, postprandial bloating - Different forms of SIBO can coexist 6. Hydrogen Sulfide-Dominant SIBO - H₂S and intestinal motility- H₂S and intestinal epithelial integrity- Symptoms, associations with IBS diarrhea 7. Host Defense Mechanisms Regulating Microbial Balance- Gastric acid secretion, pancreatic enzyme activity, bile flow, intestinal motility, and more 8. Gastric Acid and Its Role in Microbial Regulation- Parietal cells secrete hydrochloric acid (HCl) and intrinsic factor - Chief cells secrete pepsinogen, conversion into pepsin in acidic conditions - Gastric acid, digestion and antimicrobial defense 9. Hypochlorhydria - Low gastric acid impairs microbial defense- Reduced acidity disrupts digestion and downstream pancreatic enzyme and bile release 10. Pancreatic Enzymes and Their Role in Microbial Regulation- Pancreatic enzymes, digestion, antimicrobial activity - Enzymes modify chyme to limit fermentable substrates that fuel microbial proliferation 11. Intestinal Motility - Coordinated contractions propel food, microbes, and waste through the GI tract - The enteric nervous system (ENS) and gut motility- The myenteric plexus controls peristalsis, while the submucosal plexus regulates secretion and absorption 12. Conclusion- Intestinal methanogen overgrowth (IMO), hydrogen-dominant SIBO, and hydrogen sulfide-dominant SIBO - Overlap in symptoms and microbial interactions - Host defense mechanisms, including gastric acid, pancreatic enzymes, and motility - Intestinal motility and ENS function in microbial homeostasis - Hydrogen sulfide as a gasotransmitterThank you to our episode sponsors:1. Shop Fresh Press Farms'Peach Cider Vinegar at Sprouts locations nationwide, and check out their full collection here. 2. Shop CYLN's full skincare line here.Get Chloe's Book Today! "75 Gut-Healing Strategies & Biohacks" Follow Chloe on Instagram @synthesisofwellnessVisit synthesisofwellness.com
In this episode, we detail the gut-immune axis, focusing on the protective mucus layer, antimicrobial peptides, and the role of lactoferrin in intestinal health. We discuss endogenous secretions and how lactoferrin functions as an antimicrobial, immunomodulatory, and iron-regulating glycoprotein, influencing microbial balance and intestinal barrier integrity. Additionally, we highlight lactoferrin testing and levels. We finish by going through supplementation as a phenomenal tool.Topics: 1. The Intestinal Barrier & Mucus Layer- The intestinal lumen, microbiome, mucus layer, epithelial cells, and lamina propria.- The mucus layer: antimicrobial peptides (AMPs) and secretory IgA (sIgA).- Intestinal epithelial cells including goblet cells and enterocytes.2. Immune Cells in the Gut- The lamina propria.- Macrophages, dendritic cells, neutrophils, B cells, T cells, mast cells, and more.3. Antimicrobial Peptides & Secretory IgA- AMPs and sIgA in the mucus layer regulate microbial balance and prevent pathogen adhesion.4. Lactoferrin: Functions & Sources- Iron-binding glycoprotein, antimicrobial and immunomodulatory properties.- Found in mucosal secretions, colostrum, and more.- Intestinal epithelial cells (IECs) and neutrophils.5. Lactoferrin's Role in Intestinal Health- Regulates iron absorption via lactoferrin receptors (LFR)on enterocytes.- Sequesters free iron, limiting bacterial growth, bacteriostatic.- Destabilizes bacterial membranes, cell lysis, bactericidal.6. Lactoferrin Levels- Neutrophils release lactoferrin during infection,increasing its presence in the large intestine.- Fecal lactoferrin levels.- Fecal calprotectin levels.7. Lactoferrin & Candida Overgrowth- Fungal membranes.- Aiding in the inhibition of biofilm formation.8. Lactoferrin Supplementation & Benefits- Supports gut microbiota, epithelial integrity, and immunemodulation.- Bovine colostrum and as a supplement.9. Conclusion- The intestinal mucus layer is key to gut barrier function.- Lactoferrin plays a central role in microbial regulationand immune defense.Thank you to our episode sponsors:1. Shop Fresh Press Farms'Peach Cider Vinegar at Sprouts locations nationwide, and check out their full collection here. 2. Shop CYLN's full skincare line here.Get Chloe's Book Today! "75 Gut-Healing Strategies & Biohacks" Follow Chloe on Instagram @synthesisofwellnessVisit synthesisofwellness.com
In this episode, we detail the bidirectional relationship between the HPA axis and the intestinal barrier, illustrating how HPA axis dysfunction and cortisol dysregulation can impact intestinal permeability. We further discuss how intestinal dysbiosis can contribute to HPA axis overstimulation. Lastly, we detail some of the roles of short-chain fatty acids (SCFAs) and secretory IgA (sIgA) in this bidirectional relationship.Topics: 1. Overview of the HPA Axis and Gut Bidirectional Relationship- Chronic HPA activation, intestinal permeability, mucosalimmunity, and microbiome composition. 2. Components and Function of the HPA Axis- Hypothalamus, pituitary gland, and adrenal glands. - Stress signals from the amygdala and prefrontal cortex, CRH release from the hypothalamus. - CRH stimulates ACTH release from the pituitary.- Cortisol production and release. 3. Structure of the Intestinal Epithelial Barrier- The gut barrier consists of the intestinal lumen, microbiome, mucus layer, epithelial cells, and lamina propria. - Specialized epithelial cells: goblet cells, enterocytes, enteroendocrine cells, and more.- The lamina propria contains immune cells, blood vessels, and lymphatics, supported by a smooth muscle layer. 4. Intestinal Permeability and Tight Junction Regulation- Transport across the intestinal epithelium: transcellular and paracellular pathways. - Nutrient absorption and selective permeability. - Tight junction proteins, such as occludin and claudins. 5. Cortisol's Impact on Gut Barrier Integrity- Cortisol can cross the intestinal microvascular endothelium and enter the lamina propria. - Chronic cortisol exposure and intestinal barrier function.- Increased permeability allows luminal antigens and bacterial endotoxins (e.g., LPS) to infiltrate the lamina propria. 6. Secretory IgA (sIgA) and Gut Immune Function - Chronic cortisol exposure can reduce sIgA levels, weakening mucosal immunity. - sIgA neutralizes pathogens, prevents microbial adhesion, and more. - Lower sIgA levels increase susceptibility to dysbiosis and infections. 7. CRH and Its Role in Intestinal Permeability- CRH is produced in the hypothalamus and also peripherally.- CRH can stimulate mast cells, triggering histamine and inflammatory mediator release. - Mast cell activation can increase gut permeability. 8. Microbiome's Influence on HPA Axis Regulation- Dysbiosis can disrupt HPA axis function.- IBS and sustained HPA activation.- Dysbiosis reduces SCFA production. - SCFAs, particularly butyrate, support colonocyte health, tight junction integrity, and anti-inflammatory pathways. - Inflammation and HPA axis dysfunction. 9. Root Cause Approach & Closing- Chronic cortisol exposure, CRH signaling, and gut barrier dysfunction. - The microbiome influences stress response and HPA axis activity. - Roles of SCFAs, sIgA, and tight junction proteins.- Addressing gut dysbiosis and GI-derived inflammation can support HPA axis regulation. - Root cause approach.Thank you to our episode sponsors:1. Shop Fresh Press Farms'Peach Cider Vinegar at Sprouts locations nationwide, and check out their full collection here. 2. Shop the Scalp Cleanser and Scalp Essence from T Stem Care. 3. Shop Ulyana Organics'Tallow Wild Yam Cream, and use code CHLOE10 10% off your order.Get Chloe's Book Today! "75 Gut-Healing Strategies & Biohacks" Follow Chloe on Instagram @synthesisofwellnessVisit synthesisofwellness.com
In this episode, we go through the pathophysiology of hydrogen sulfide (H₂S)-dominant small intestinal bacterial overgrowth (SIBO), focusing on its role in intestinal motility disruption, microbial dysbiosis, and epithelial barrier dysfunction. We detail the interplay between the enteric nervous system (ENS), interstitial cells of Cajal (ICCs), and migrating motor complex (MMC) in regulating small intestinal transit and how excess H₂S modulates smooth muscle activity, inflammatory signaling, and gastrointestinal transit time. Lastly, we detail symptoms and related SIBO subtypes.Topics:1. Introduction to Hydrogen Sulfide Dominant SIBO - Overview of hydrogen sulfide (H₂S) SIBO - Sulfate as an electron acceptor - H₂S production, sulfur-containing amino acids - Physiological roles of H₂S vs. in excess 2. Hydrogen Sulfide and Gastrointestinal Motility - Impact on transit time - Association with IBS-like symptoms, diarrhea 3. Intestinal Motility and Regulation - Role of the ENS, ICCs, smooth muscle cells, and neurotransmitters in motility 4. The Enteric Nervous System (ENS) and Gut Motility - ENS as the "second brain" and its control over digestion - Myenteric and submucosal plexuses: regulation of peristalsis, secretion, blood flow - Gut-brain axis involvement via vagus nerve signaling - ICCs as pacemaker cells generating slow-wave electrical activity - MMC's role in clearing debris and bacteria 5. Dysregulated Motility and SIBO Development 6. Overlapping Symptoms Across SIBO Subtypes - Shared symptoms - Hydrogen and H₂S-dominant SIBO vs. IMO 7. Gastric Acid and Its Role - HCl secretion by parietal cells and its role in digestion and microbial defense - Hypochlorhydria and bacterial overgrowth 8. Conclusion - Root cause approach.Thank you to our episode sponsors:1. Check out Ulyana Organics'Tallow Wild Yam Cream and Healing Facial Oil, and use code CHLOE10 10% off your order.2. Shop the Scalp Cleanser and Scalp Essence from T Stem Care. Get Chloe's Book Today! "75 Gut-Healing Strategies & Biohacks" Follow Chloe on Instagram @synthesisofwellnessVisit synthesisofwellness.com
In this episode, we discuss the relationship between the intestinal mucus layer and secretory immunoglobulin A (sIgA), highlighting both as key components of the gut's first line of defense against pathogens, toxins, and environmental stressors. We detail how dysbiosis and microbial overgrowths can impact sIgA levels and can disrupt mucus integrity and function, contributing to increased intestinal permeability and inflammation. We then examine several additional factors influencing sIgA production. Topics: 1. Introduction: Importance of Secretory IgA and Intestinal Mucus - Intestinal epithelial lining and its protective mucus layer - Focus on intestinal dysbiosis and sIgA 2. Anatomy of the Gastrointestinal Lining - Intestinal lumen, microbiome, mucus layer, epithelial cells, tight junctions, lamina propria, and smooth muscle - Role of goblet cells in mucus secretion and epithelial integrity 3. Role of the Protective Mucus Layer - Dual function of the mucus layer as a physical and biochemical barrier - Composition of mucus: mucins, antimicrobial peptides, immunoglobulins 4. Goblet Cells and Mucin Production - Mechanisms of mucin secretion and hydration - Variability in goblet cell density across the intestinal tract - Dysfunctions linked to conditions like IBD and IBS 5. Microbial Interactions with Mucins - Glycan structures in mucins - Roles of commensal and pathogenic bacteria in mucin degradation and mucus turnover - Importance of regulated mucus production for gut homeostasis 6. Dysbiosis and Mucus Degradation - Excessive mucin degradation leading to thinning of the mucus layer - Consequences of increased intestinal permeability and inflammation - Role of dysbiosis in disrupting the mucus layer 7. Overview of Secretory IgA - Introduction to secretory IgA (sIgA) and its role in mucosal immunity - Mechanisms of immune exclusion and pathogen neutralization 8. Production and Function of Secretory IgA - sIgA production by plasma cells in the lamina propria - Translocation of IgA through epithelial cells into the gut lumen - sIgA binding to mucins and its immunological role in the mucus layer 9. Factors Affecting Secretory IgA Levels - Possible contributing factors to low sIgA levels - Implications of low and high sIgA levels for immune defense and intestinal health 10. Interactions Between Secretory IgA and Dysbiosis - Cyclical relationship between low sIgA, dysbiosis, and infections - Examples of overgrowths, like candida, disrupting sIgA - Importance of addressing immune dysregulation to break the cycle 11. Bioindividual Root Cause Approach - Factors to consider Thank you to our episode sponsors: 1. Shop Fresh Press Farms' Peach Cider Vinegar at Sprouts locations nationwide, and check out their full collection here. 2. Check out Spectrum Sciences' Serums and The Vitamin B12 Protocol here, and use code CHLOE15 for 15% off. Get Chloe's Book Today! "75 Gut-Healing Strategies & Biohacks" Follow Chloe on Instagram @synthesisofwellness Visit synthesisofwellness.com
In this episode, we go through the gut-immune axis and mycobiome, focusing on the mechanisms underlying intestinal fungal overgrowth. We highlight the roles of the intestinal mucus layer, intestinal epithelial cells, lamina propria, and secretory immunoglobulin A (sIgA) in maintaining homeostasis and defending against fungal overgrowth. We also detail the pathogenic transition of Candida from its commensal yeast form to an invasive hyphal state, as well as the resulting disruptions to immune function. Topics 1. Introduction to the Gut-Immune Axis and Mycobiome - The gut-immune axis and mycobiome. - Exploring intestinal fungal overgrowth. 2. Overview of the Gastrointestinal Lining - Anatomy of the intestinal lining: lumen and microbiome, mucus layer, epithelial monolayer, and lamina propria. - The dual role of the mucus layer: physical and biochemical barrier. - Composition of mucus. 3. Goblet Cells and Mucus Production - Goblet cells as critical producers of mucin glycoproteins and their role in mucus barrier integrity. - Signals for mucin release, such as mechanical stress. 4. The Lamina Propria and Gut-Associated Lymphoid Tissue (GALT) - Layers beneath the epithelium - Peyer's patches, mesenteric lymph nodes, isolated lymphoid follicles, and intraepithelial lymphocytes. - Immune tolerance and defense against pathogens. 5. Peyer's Patches - Positioning of Peyer's patches. - The transition of microbial density from the small intestine to the colon and its immune implications. 6. Immune Cells in the Lamina Propria - Overview of macrophages, dendritic cells, mast cells, plasma cells, and lymphocytes. - Their role in maintaining immunological equilibrium in the gut. 7. The Mycobiome and Pathogenic Transition of Candida - Candida's transition from yeast to hyphal form. - Regulation of Candida by microbial competition and immune surveillance. - Factors promoting pathogenicity: immune suppression, lacking beneficial microbes, more. 8. Virulence Factors of Candida - Secretion of hydrolytic enzymes such as SAPs and phospholipases. - Degradation of sIgA and its consequences for immune evasion. - Adhesins that facilitate fungal binding. 9. Intestinal Barrier Disruption by Candida - Penetration of the mucus layer and epithelial barrier by Candida hyphae. - Disruption of tight junctions and the resulting increased intestinal permeability. - Inflammatory cascade triggered by fungal antigens and microbial toxins entering the lamina propria. 10. Biofilm Formation - The role of biofilms in shielding fungal cells from immune attack and antifungal treatments. 11. Small Intestinal Fungal Overgrowth (SIFO) - Intestinal dysmotility, low stomach acid, immunocompromise. - Possible symptoms of SIFO. - Discussion of the overlap between SIFO, SIBO, histamine intolerance, and other gastrointestinal conditions. - Root cause. Thank you to our episode sponsors: 1. Shop Fresh Press Farms' Peach Cider Vinegar at Sprouts locations nationwide, and check out their full collection here. 2. Check out Ulyana Organics' Tallow Wild Yam Cream and Healing Facial Oil, and use code CHLOE10 10% off your order. 3. Check out Spectrum Sciences' Serums and The Vitamin B12 Protocol here, and use code CHLOE15 for 15% off. Get Chloe's Book Today! "75 Gut-Healing Strategies & Biohacks" Follow Chloe on Instagram @synthesisofwellness Visit synthesisofwellness.com
We are honored to be joined today by Chris Soppet. Chris is the founder of Spectrum Sciences, a health and wellness brand with the vision of bringing the healing power of vitamin B12 to the greatest number of folks out there. He received his inspiration from his son, who recovered from autism in large part due to the remarkable power of vitamin B12. Chris shares this hope through a very tangible means – The Vitamin B12 Protocol. After using various other B12 products on the market, he was disappointed by the inconsistency and lack of transparency. As a chemist and certified health coach, he was uniquely positioned to take matters into his own hands. Launched in the Fall of 2024, Spectrum Sciences aims to deliver the most effective, safe, and transparently labeled option in transdermal delivery of nutrients. Topics: 1. Chris' Journey and Developing Spectrum Sciences - Chris Soppet's background and the inspiration for founding Spectrum Sciences. 2. Brief Overview of The Vitamin B12 Protocol 3. Brief Overview of Methylation - The importance of methylation for cellular function and overall health. 4. Background - The role of the thyroid in supporting methylation and B12 utilization, including testing, nutrients, and understanding a basic thyroid panel. - The thyroid's role in activating riboflavin (B2) and other cofactors, and how activated riboflavin is used. - How to support riboflavin status through dietary intake. - The folate cycle and how activated riboflavin supports the MTHFR enzyme to yield active folate. - The interaction of B12 and B9 in converting homocysteine to methionine and producing SAM, the universal methylator. 5. The Organic Acids Test - Explanation of the test and its insights. 6. Topical Serums - Benefits of topical delivery. - Spectrum Sciences' products that support thyroid function. - Spectrum Sciences' products that support riboflavin activation with thyroid assistance. - Spectrum Sciences' activated B12. - Remarkable transdermal delivery through microemulsion technology. - How to use the topicals. 7. Spectrum Sciences LLC - Resources for learning about Spectrum Sciences, The Vitamin B12 Protocol, and topical serums. 8. Connecting with Chris Soppet - Social media and website links to learn more about Chris and Spectrum Sciences. Check out Spectrum Sciences and The Vitamin B12 Protocol at spectrum-sciences.net, and use code CHLOE15 for 15% off. Follow Spectrum Sciences on Instagram @spectrumscience1
In this episode, we explore some of the neural and cellular mechanisms underlying gastrointestinal motility, emphasizing the roles of the enteric nervous system (ENS) and interstitial cells of Cajal (ICCs) in coordinating smooth muscle contractions throughout the digestive tract. We detail disruptions in motility seen in dysautonomia, intestinal methanogen overgrowth (IMO), and small intestinal bacterial overgrowth (SIBO). And, we finish with a brief discussion on the impacts of low stomach acid. Topics: 1. Introduction to Gastrointestinal Motility - Coordinated contractions of smooth muscle that move food, liquid, and waste through the gastrointestinal (GI) tract. - Neural, hormonal, and microbial factors. - Intestinal methanogen overgrowth (IMO) and small intestinal bacterial overgrowth (SIBO). 2. Neural Regulation of Gut Motility - The enteric nervous system (ENS) embedded in the walls of the GI tract. - The ENS, central nervous system (CNS), and vagus nerve. - The myenteric plexus governs motor control and peristalsis, while the submucosal plexus regulates secretion, absorption, and blood flow. - Stress and motility 3. Cellular Regulation of Gut Motility - Interstitial cells of Cajal (ICCs), pacemaker cells located near smooth muscle cells. - ICCs generate slow-wave electrical activity. - Acetylcholine enhances smooth muscle contraction. 4. Dysmotility and Potential Contributing Factors - Impaired ICC function, smooth muscle dysfunction, nervous system dysfunction. - Dysautonomia, a dysfunction of the autonomic nervous system. - External stressors, hormonal imbalances, and more. 5. Intestinal Methanogen Overgrowth (IMO) - Characterized by an overgrowth of methane-producing archaea in the intestines. - Excess methane production affects smooth muscle activity and can slow intestinal transit. - Symptoms such as bloating, abdominal discomfort, constipation. - Breath testing. 6. Small Intestinal Bacterial Overgrowth (SIBO) - Excessive bacterial colonization in the small intestine, disrupting normal gut function. - Protective mechanisms including stomach acid and intestinal motility. - Low stomach acid (hypochlorhydria) reduces the acidic environment needed to inhibit bacterial colonization. - Bloating, diarrhea/constipation, and more. 7. Overlap Between IMO and SIBO - Excess hydrogen produced in SIBO can serve as a substrate for methane production in IMO. - Can exacerbate each other, creating a loop that worsens symptoms. 8. Addressing Dysmotility and Overgrowth - Determining the root cause of dysmotility. - Dysmotility can involve multiple regions of the GI tract. Thank you to our episode sponsors: 1. Shop Fresh Press Farms' Peach Cider Vinegar at Sprouts locations nationwide, and check out their full collection here. 2. Check out Ulyana Organics' Tallow Wild Yam Cream and Healing Facial Oil, and use code CHLOE10 10% off your order. Thanks for tuning in! Get Chloe's Book Today! "75 Gut-Healing Strategies & Biohacks" Follow Chloe on Instagram @synthesisofwellness Follow Chloe on TikTok @chloe_c_porter Visit synthesisofwellness.com to purchase products, subscribe to our mailing list, and more!
In this episode, we detail the gut's intricate immune defenses, emphasizing the role of mast cells in inflammation and immune signaling. We extend this conversation to Mast Cell Activation Syndrome briefly going through potential triggers, tests, and symptoms. Finally, we examine the bidirectional relationship between MCAS and Small Intestinal Bacterial Overgrowth (SIBO), illustrating how mast cell mediators and microbial byproducts can drive a cycle of chronic inflammation and increased intestinal permeability. Topics: 1. Introduction to the Gut-Immune Axis - Focus: mast cells, small intestinal bacterial overgrowth (SIBO), and mast cell activation syndrome (MCAS). 2. Anatomy of the Gastrointestinal Lining - Intestinal lumen, microbiome, mucus layer, epithelial monolayer, tight junctions, and lamina propria. - Role of the smooth muscle and deeper layers supporting the mucosal structures. 3. Gut-Associated Lymphoid Tissue (GALT) - Peyer's patches, mesenteric lymph nodes (MLNs), isolated lymphoid follicles (ILFs), intraepithelial lymphocytes (IELs), and dispersed immune cells in the lamina propria. - Function of GALT. 4. Peyer's Patches and Antigen Exposure - Location and function of Peyer's patches. - Role in antigen sampling and processing. 5. Immune Cells in the Lamina Propria - Overview of interspersed immune cell populations. 6. Focus on Mast Cells - Key roles of mast cells in the gut's innate immune system. - Locations with a focus on the lamina propria. 7. Mast Cell Mediators - Histamine - Tryptase: tissue remodeling, impact on intestinal permeability. - Cytokines, prostaglandins, and leukotrienes. 8. Mast Cell Activation Syndrome (MCAS) - Overactivation of mast cells and excessive release of inflammatory mediators. - Effects on gut barrier function. - Pathological behavior due to altered activation thresholds, receptor expression, and tissue environment changes (not resulting from an increased number of mast cells). 9. MCAS Symptoms and Systemic Effects - Abdominal pain, cramping, bloating, diarrhea, and nausea. - Systemic symptoms: skin reactions, respiratory and cardiovascular effects, neurological impacts. 10. Triggers and Conditions Associated with MCAS - Environmental toxins, infections, stress, chemical exposures. - Hypermobile Ehlers-Danlos syndrome (hEDS), dysautonomia (e.g., POTS). 11. MCAS Testing - Testing limitations: variability in mediator release and transient nature of mast cell degranulation. - Serum tryptase, urinary N-methylhistamine, plasma heparin levels, specific cytokines like IL-6, and more. 12. Small Intestinal Bacterial Overgrowth (SIBO) - Overview of SIBO. - Slow motility, low stomach acid. 13. Interaction Between SIBO and Mast Cells - SIBO-induced mast cell activation. - Cycle of inflammation, increased intestinal permeability, and gut dysfunction. 14. Conclusion - Recap of the intestinal anatomy and immune cell focus. - MCAS triggers, symptoms, and testing. - Connections between SIBO and MCAS, emphasizing an inflammatory cycle. Thank you to our episode sponsor: 1. Check out Ulyana Organics' Tallow Wild Yam Cream and Healing Facial Oil, and use code CHLOE10 10% off your order. Thanks for tuning in! Get Chloe's Book Today! "75 Gut-Healing Strategies & Biohacks" Follow Chloe on Instagram @synthesisofwellness Follow Chloe on TikTok @chloe_c_porter Visit synthesisofwellness.com to purchase products, subscribe to our mailing list, and more!
In this episode, we go through the role of histamine within the gastrointestinal tract, exploring its regulation of gastric acid secretion and impact on intestinal motility. We examine how histamine-producing enterochromaffin-like (ECL) cells, mast cells, and gut microbiota contribute to histaminergic signaling, and how enzymatic degradation via diamine oxidase (DAO) maintains homeostasis. Additionally, we discuss histamine intolerance, highlighting the impacts of dysbiosis (as well as SIBO or small intestinal bacterial overgrowth) and intestinal barrier dysfunction on DAO insufficiency and histamine accumulation. Topics: 1. Introduction to Histamine & the Gastrointestinal Tract Role in digestion and motility Impact of intestinal conditions such as SIBO 2. Histamine as a Biogenic Amine Definition and classification L-histidine and histidine decarboxylase (HDC) Presence in various tissues, including the GI tract 3. Sources of Histamine in the Gut Enterochromaffin-like (ECL) cells in the stomach Mast cells in the lamina propria Histamine-producing gut bacteria 4. Histamine's Role in Digestion & Gastric Acid Secretion Interaction with parietal cells in the stomach Activation of proton pumps via H₂ receptor binding Maintenance of acidic pH for digestion Importance of proper pH within the stomach for protecting against pathogenic microbes Impact on downstream gut microbiota balance (e.g., SIBO) 5. Histamine & Intestinal Motility Modulation of gut motility via H₁ and H₃ receptors H₁ receptor activation leading to contractions (diarrhea, cramping) H₃ receptor-mediated inhibition of neurotransmitter release (bloating, slow motility) 6. Histamine Intolerance: Definition & Mechanisms Diamine Oxidase and histamine breakdown DAO secretion by enterocytes (intestinal epithelial cells) Enzymatic breakdown of histamine before absorption 7. Factors That Can Affect DAO Activity Genetic Polymorphisms: AOC1 gene, reduced DAO expression Cofactor Deficiencies: Copper, vitamin B6, and vitamin C as essential DAO cofactors Importance of intestinal lining integrity for DAO production Conditions leading to or associated with enterocyte damage Intestinal dysbiosis and histamine accumulation Intestinal dysbiosis & epithelial/enterocyte damage Suppression of DAO production due to epithelial dysfunction Small Intestinal Bacterial Overgrowth (SIBO) & Histamine 8. Symptoms of Histamine Intolerance Gastrointestinal symptoms (bloating, gas, constipation, diarrhea, acid reflux...) Systemic symptoms (dizziness, flushing, migraines...) 9. Identifying Potential Root Causes Tools and supplements Bioindividual approach Thanks for tuning in! Get Chloe's Book Today! "75 Gut-Healing Strategies & Biohacks" Follow Chloe on Instagram @synthesisofwellness Follow Chloe on TikTok @chloe_c_porter Visit synthesisofwellness.com to purchase products, subscribe to our mailing list, and more!
In this episode, we discuss the anatomy and physiology of the stomach and its unique epithelial structure. We dive into the pathophysiological implications of conditions such as Helicobacter pylori infection and hypochlorhydria, examining how they disrupt gastric function and microbial balance. Finally, we connect these concepts to the small intestine, discussing possible downstream effects of altered gastric acid secretion, focusing on microbial colonization and contributions to broader gastrointestinal health challenges, such as small intestinal bacterial overgrowth (SIBO). Topics: 1. Introduction Highlighting the stomach and anatomy. Discussion on H. pylori. Interplay between the stomach and small intestines, focusing on microbial balance and overgrowth. 2. Anatomy of the Stomach The stomach as a hollow, muscular organ located between the esophagus and small intestine. Anatomical regions: cardia, fundus, body, and pylorus. Layers of gastric mucosa: epithelial lining, lamina propria, and thin band of smooth muscle. 3. Epithelial Lining and Specialized Cells Mucous cells: Location within the surface epithelium and gastric pits. Secretion of viscous mucus containing mucins, forming a protective barrier. Role in shielding the epithelial lining from acidic gastric juices and creating a neutral microenvironment. Parietal cells: Location in the gastric glands, predominantly in the fundus and body. Secretion of hydrochloric acid (HCl) for digestion and defense. Production of intrinsic factor and vitamin B12 absorption. Chief cells: Secretion of pepsinogen, converted to pepsin for protein digestion. Production of gastric lipase for lipid digestion. Enteroendocrine cells: Hormone secretion. Gastrin, acid secretion and gastric motility. 4. Protection and Functionality of Gastric Mucosa Physical and chemical barriers. Acidic environment maintained by parietal cells for enzymatic activity and pathogen defense. Coordination of cell functions for efficient digestion and protection. 5. Transition from Stomach to Small Intestine Anatomical junction marked by the pyloric sphincter. Balancing the acidic environment of the stomach with the intestinal conditions. Differences between stomach and intestinal epithelial barriers. 6. Microbial Communities in the Stomach and Intestines Limited microbial diversity in the stomach due to its low pH. Acid-resistant populations such as H. pylori and their impact on microbial diversity. H. pylori , chronic gastritis, ulcers, and gastric lining atrophy. 7. H. Pylori Infection and Symptoms Symptoms, manifestations. 8. Low Stomach Acid (Hypochlorhydria) The role of stress, aging, and infections. Downstream effects, particularly in the development of small intestinal bacterial overgrowth (SIBO). 9. Small Intestinal Bacterial Overgrowth (SIBO) Definition and microbial population thresholds. Protective mechanisms regulating bacterial density in the small intestine. Role of low stomach acid in bacterial colonization and overgrowth. Consequences of SIBO, including symptoms and metabolic activity of overgrown bacteria. 10. Conclusion Root cause analysis. Thank you to our episode sponsor: 1. Check out Ulyana Organics' Tallow Wild Yam Cream and Healing Facial Oil, and use code CHLOE10 10% off your order. Thanks for tuning in! Get Chloe's Book Today! "75 Gut-Healing Strategies & Biohacks" Follow Chloe on Instagram @synthesisofwellness Follow Chloe on TikTok @chloe_c_porter Visit synthesisofwellness.com to purchase products, subscribe to our mailing list, and more!
This episode explores three top topics from episodes in 2024: the mucus layer's protective role in maintaining gut barrier integrity, the bidirectional relationship between the microbiota-gut-brain axis and the hypothalamic-pituitary-adrenal (HPA) axis, and the importance of methylation and the MTHFR gene. During the first topic of discussion, we detail ways to support the intestinal mucus layer; as we approach the third topic of discussion, we highlight the impact of MTHFR enzyme activity on methylation, emphasizing its importance for many cellular processes. Topics: 1. Introduction: 3 Top Topics of 2024 2. The Protective Mucus Layer Over the Intestinal Epithelial Lining The intestinal barrier: intestinal lumen, microbiome, mucus layer, epithelial cells, lamina propria, and more. The mucus layer functions as both a physical and biochemical barrier. Goblet cells secrete mucins, which form the gel-like mucus layer, critical for maintaining barrier integrity. Dysregulation of mucus turnover can lead to increased intestinal permeability and inflammation. Supporting mucus levels: SCFAs, N-acetylglucosamine (NAG). Plant-derived mucilage and sources. 3. HPA Axis and Microbiota-Gut-Brain Interactions The HPA axis and the stress response. Microbiota-gut-brain axis. SCFAs produced by gut bacteria can influence gut barrier integrity and stress resilience. Dysbiosis disrupts SCFA production, weakens the gut barrier, and triggers inflammation. Dysbiosis and HPA axis dysregulation. Chronic cortisol secretion and secretory IgA. 4. The MTHFR Gene and Methylation Methylation involves transferring methyl groups. The synthesis of SAMe, the universal methyl donor: methionine and ATP with active folate (L-methylfolate) as a precursor. MTHFR gene polymorphisms reduce enzyme efficiency, impairing methylation and affecting DNA stability, neurotransmitter synthesis, and detoxification. Strategies, supplements, and more to support methylation. Thank you to our episode sponsor: 1. Check out Ulyana Organics' Tallow Wild Yam Cream and Healing Facial Oil, and use code CHLOE10 10% off your order. Thanks for tuning in! Get Chloe's Book Today! "75 Gut-Healing Strategies & Biohacks" Follow Chloe on Instagram @synthesisofwellness Follow Chloe on TikTok @chloe_c_porter Visit synthesisofwellness.com to purchase products, subscribe to our mailing list, and more! --- Support this podcast: https://podcasters.spotify.com/pod/show/chloe-porter6/support
In this episode, we go through the interplay between the oral microbiome, the intestinal microbiome, and brain health, focusing on the mechanisms through which dysbiosis in these ecosystems influences neuroinflammation and central nervous system function. We explore the molecular pathways by which bacterial metabolites and inflammatory mediators from the oral and intestinal microbiota can disrupt the blood-brain barrier, activate glial cells, and impair neuronal processes. We also go through symptoms associated with microbial imbalance as well as oral health practices. Topics: 1. Introduction Focus on neuroinflammation, oral microbiome, and intestinal microbiome Oral-gut-brain axis. 2. Neuroinflammation Overview Inflammatory response within the CNS. Key cell types: Endothelial cells, neurons, glial cells (microglia, astrocytes, oligodendrocytes). Acute vs. chronic neuroinflammation and their implications. 3. Cellular Mechanisms Microglial activation and its role in cytokine and ROS production. Astrocytic contributions to neuroinflammation. 4. Effects of Neuroinflammation on the Brain Mitochondrial dysfunction and energy deficits in neurons. Blood-brain barrier (BBB) disruption and immune cell infiltration. Impacts on synaptic function and neuronal communication. Potential contributors - environmental toxins, sleep deprivation, microbial dysbiosis, infections, and more. 5. Role of the Intestinal Microbiome in Neuroinflammation Dysbiosis and the release of LPS by Gram-negative bacteria. LPS transport across the intestinal barrier (transcellular and paracellular pathways). Systemic inflammation and its impact on BBB integrity and CNS inflammation. 6. The Oral Microbiome and Brain Health Direct effects of oral dysbiosis on systemic inflammation. Indirect effects through oral-gut communication and intestinal dysbiosis. 7. Trigeminal Nerve and Oral Dysbiosis Role of the trigeminal nerve in sensory transmission. Interaction of oral bacteria with the trigeminal nerve via TLRs and LPS recognition. 8. Symptoms and Effects of Oral Dysbiosis Common symptoms Systemic effects on the intestinal microbiome and brain health. 9. Lifestyle Practices Oral health practices. 10. Conclusion Summary of the oral-gut-brain axis and its role in neuroinflammation. Importance of oral health in reducing inflammatory burdens. Thank you to our episode sponsor: 1. Check out Ulyana Organics' Tallow Wild Yam Cream and Healing Facial Oil, and use code CHLOE10 10% off your order. Thanks for tuning in! Get Chloe's Book Today! "75 Gut-Healing Strategies & Biohacks" Follow Chloe on Instagram @synthesisofwellness Follow Chloe on TikTok @chloe_c_porter Visit synthesisofwellness.com to purchase products, subscribe to our mailing list, and more! --- Support this podcast: https://podcasters.spotify.com/pod/show/chloe-porter6/support
In this episode, we dive into the interplay between the gut microbiota (their postbiotics) and the immune system. We will examine how gut microbiome-derived metabolites, such as short-chain fatty acids (SCFAs) and polyphenol metabolites, influence immune function by modulating key cellular and molecular pathways within the intestinal mucosa. Additionally, we discuss strategies for supporting butyrate production and optimizing microbiome health to foster a balanced and resilient gut-immune axis. Topics: 1. Introduction Overview of the gut-immune axis. Importance of gut microbiome-derived metabolites in supporting immune function. 2. The Intestinal Barrier Layers of the intestinal wall Focus on mucosa, specifically the epithelium and lamina propria. 3. Structure of the Intestinal Layers The intestinal lumen, mucus layer, epithelium (with tight junctions), and lamina propria. Importance of the lamina propria as a hub for immune responses and structural integrity. 4. Cellular and Structural Components of the Lamina Propria Extracellular matrix (ECM): structural support. Fibroblasts and myofibroblasts. Lymphatic vessels: immune cell transport, linking mucosal and systemic immune systems. 5. Immune Cells in the Lamina Propria T cells: immune tolerance, regulatory T cells (Tregs). B cells: Secretory immunoglobulin A (sIgA). Dendritic cells: antigen sampling and presentation. Macrophages: pathogen clearance. Mast cells 6. Role of Secretory Immunoglobulin A (sIgA) Functions as a first-line defense in the intestinal mucus layer. Neutralizes pathogens, prevents epithelial adhesion. 7. Postbiotics Overview Bioactive compounds produced by gut microbiota. Examples: short-chain fatty acids (SCFAs) 8. Short-Chain Fatty Acids (SCFAs) and Their Functions Influence on Treg cells in the lamina propria, promoting immune tolerance. Butyrate also as an energy source for epithelial cells. 9. Supporting Butyrate Production Microbiome optimization to enhance beneficial butyrate-producing microbes. Use of prebiotics: resistant starch, soluble fibers, and polyphenols. Supplementation with sodium butyrate as an additional tool. 10. Other Postbiotics Antimicrobial peptides produced by beneficial microbes. Complex carbohydrates produced by beneficial microbes and can act as prebiotics. Polyphenol metabolites: Gut microbiota biotransforms polyphenols into bioactive metabolites with increased bioavailability. 11. Specific Polyphenols Examples: resveratrol, quercetin, and ellagitannins. Effects on intestinal barrier function, inflammation, and immune cell populations. "75 Gut-Healing Strategies & Biohacks" Follow Chloe on Instagram @synthesisofwellness Follow Chloe on TikTok @chloe_c_porter Visit synthesisofwellness.com --- Support this podcast: https://podcasters.spotify.com/pod/show/chloe-porter6/support
In this episode, we explore the intricate role of the vagus nerve as a central regulator within the microbiota-gut-brain (MGB) axis, examining its neuroanatomical structure, signaling mechanisms, and interactions with microbial metabolites and immune pathways. We discuss how vagal afferent fibers relay sensory input from the gut to the brain, including signals mediated by short-chain fatty acids (SCFAs) and gut-derived hormones, and how efferent fibers modulate gut motility, intestinal barrier integrity, and inflammation through the cholinergic anti-inflammatory pathway. Finally, we explore vagal dysfunction as well as associated conditions and symptoms, and we touch on just a few potential root causes. Topics: 1. Introduction Focus on the vagus nerve's role in the microbiota-gut-brain (MGB) axis. Bidirectional communication between the brain and microbiota. Overview of communication pathways: neural (e.g., vagus nerve), endocrine (e.g., HPA axis), immune (e.g., cytokines), and metabolic (e.g., SCFAs). 2. Overview of the Nervous System The CNS includes the brain and spinal cord - control centers for the body. The peripheral nervous system extends beyond the CNS The peripheral nervous system is divided into the somatic nervous system and the autonomic nervous system. 3. Autonomic Nervous System (ANS) and Subdivisions Sympathetic Nervous System (SNS) Parasympathetic Nervous System (PNS) Enteric Nervous System (ENS) 4. The Vagus Nerve and Role in the PNS Principal component of the parasympathetic nervous system. Governs "rest-and-digest" activities Contains both afferent (80%) and efferent (20%) fibers. 5. Vagus Nerve Anatomy Fibers originate at the base of the skull and extend into the gut wall. Fibers distributed throughout the mucosa, submucosa, and beyond. Interact indirectly with gut luminal contents via specialized gut cells, including EECs and immune cells. 6. Interaction with Intestinal Cells Enteroendocrine cells (EECs) release gut hormones in response to microbial metabolites. SCFAs, such as butyrate, activate free fatty acid receptors on EECs, stimulating vagal afferents. Immune cells within the gut wall modulate vagal signals during inflammatory responses. 7. Review of Functions Sensory input (afferent fibers): Detects gut-derived signals like microbial metabolites and mechanical stretch. Motor output (efferent fibers): Regulates gut motility, secretion, immune responses, and more. 8. Impact of a Diverse Microbiome on Vagal Activity Enhanced SCFA production boosts vagal activity. SCFAs improve gut barrier integrity, reduce systemic inflammation, and assist in regulating stress responses. 9. Examples: Intestinal Barrier Function Releases acetylcholine (ACh) to modulate inflammatory pathways. Helps enhance tight junction protein expression, preserving gut barrier integrity. Helps prevent the translocation of microbial endotoxins like LPS into systemic circulation. 10. Dysfunction of the Vagus Nerve Reduced vagal tone disrupts gut homeostasis. Conditions such as IBS, IBD, chronic fatigue syndrome, anxiety, depression, and POTS. Chronic stress, infections, and dysbiosis are common contributors. 11. Root Causes 12. Tying Back to the HPA Axis Low vagal tone is associated with increased HPA axis activity. Highlighting the interplay between the gut, brain, and stress response systems. 13. Conclusion Identifying potential root causes. Contributing lifestyle factors. "75 Gut-Healing Strategies & Biohacks" Follow Chloe on Instagram @synthesisofwellness Follow Chloe on TikTok @chloe_c_porter Visit synthesisofwellness.com --- Support this podcast: https://podcasters.spotify.com/pod/show/chloe-porter6/support
In this episode, we dive into the interplay between the hypothalamic-pituitary-adrenal (HPA) axis and the microbiota-gut-brain (MGB) axis, highlighting their bidirectional communication through endocrine, immune, and neural pathways. We'll explore how gut-derived metabolites like short-chain fatty acids (SCFAs) and endotoxins like LPS influence HPA axis activity; conversely, we explore how dysregulated cortisol can impact gut barrier function, immune signaling, and more. We also discuss testing including stool analysis and DUTCH tests. Topics: 1. HPA Axis and MGB Axis Interaction The HPA axis and microbiota-gut-brain (MGB) axis are bidirectionally connected. Gut microbiota influences the HPA axis via metabolites and more 2. Overview of the HPA Axis and Cortisol Secretion Stress signals activate the hypothalamus to release CRH. Stimulates the anterior pituitary to produce ACTH. ACTH signals the adrenal cortex. The adrenal cortex releases cortisol, which binds to glucocorticoid receptors (GRs). 3. Cortisol Dysregulation Impact on Intestinal Health Intestinal lining anatomy: epithelial cells Tight junction proteins regulate nutrient trafficking and prevent pathogen entry. The mucosa contains epithelial cells, connective tissue (lamina propria), and a thin muscle layer. 4. Glucocorticoid Receptors (GRs) in the Gut GRs are intracellular receptors that modulate gene expression when activated. Cortisol binding causes GRs to translocate to the nucleus and bind DNA at GREs. GRs on epithelial cells. Modified tight junction protein expression. 5. Gut Microbiome's Role in HPA Axis Activity SCFAs, including acetate, propionate, and butyrate, produced by gut bacteria fermenting dietary fiber. SCFAs support gut integrity, reduce inflammation, and act as signaling molecules. A diverse and healthy microbiome can enhances HPA regulation via SCFAs. 6. SCFA Modulation of the HPA Axis SCFAs and cortisol. Research highlights SCFAs' ability to attenuate stress-induced cortisol increases. 7. Dysbiosis - Impact on the HPA Axis Dysbiosis reduces SCFA production, impairing gut barrier integrity and immune signaling. Dysbiosis, intestinal hyperpermeability and LPS. LPS activates inflammatory pathways. 8. Inflammation and Dysregulated HPA Activity Chronic inflammation and cortisol. Inflammatory signals from the gut exacerbate systemic and neural stress responses. 9. Symptoms of HPA Axis Dysfunction Chronic fatigue, disrupted sleep, mood disturbances... 10. Identifying Root Causes Dysbiosis, chonic infection / chronic inflammation... DUTCH Test Stool analysis Thanks for tuning in! "75 Gut-Healing Strategies & Biohacks" Follow Chloe on Instagram @synthesisofwellness Follow Chloe on TikTok @chloe_c_porter Visit synthesisofwellness.com --- Support this podcast: https://podcasters.spotify.com/pod/show/chloe-porter6/support
In this episode, we examine the bidirectional relationship between intestinal health and immune aging, emphasizing how gut barrier dysfunction and microbial imbalances accelerate systemic inflammation and diminished immune function. Furthermore, we explore the effects of aging on intestinal barrier function, focusing on the decline in tight junction integrity, reduced mucus production, and impaired epithelial renewal, all of which contribute to increased gut permeability and chronic inflammation. Thus, restoring microbial diversity, supporting mucosal defenses, and addressing gut permeability are essential strategies to break this cycle and promote healthy aging. Topics: 1. Introduction: Immune Aging - Immune aging: a decline in immune function, increasing vulnerability to infections and chronic diseases. - The intestinal microbiome influences and is shaped by immune aging. - Microbial metabolites, epithelial integrity, and immune signaling are critical. 2. Anatomical Context of the Gut and Immune Cells - Gut layers: lumen, mucus, epithelium, lamina propria, and more. - The mucus layer protects the epithelium; function can become impaired with age. - Tight junctions prevent pathogen entry, while the lamina propria houses immune cells. 3. Mucus Layer and Goblet Cells: Role and Aging Effects - Goblet cells secrete mucins that form the protective mucus layer. - Aging can reduce mucin production, weakening the mucus barrier. - A thin mucus layer increases epithelial exposure and inflammation risk. 4. Microbial Interactions with the Mucus Layer - Commensal bacteria support mucus turnover by controlled mucin degradation. - Dysbiosis can disrupt this process, thinning the mucus layer. - Reduced mucus protection increases susceptibility to pathogens. 5. Effects of Immune Aging on the Intestinal Barrier - Aging effects on T cells, B cells, and inflammatory cytokine secretion. - Cytokines disrupt tight junctions and increase permeability. - Microbial products like LPS (endotoxin) cross the barrier, fueling chronic inflammation. 6. Dysbiosis and Its Role in Immune Aging - Dysbiosis reduces SCFA production and Treg activity. - Dysbiosis promotes chronic inflammation and accelerates immune aging. 7. Microbial Translocation and Systemic Effects - A weakened barrier allows microbial products to reach immune cells. - LPS triggers inflammatory signaling, amplifying systemic inflammation. - Chronic inflammation perpetuates gut dysfunction and immune aging. 8. Conclusion - Immune aging and intestinal health are interconnected in a feedback loop. - Aging weakens the gut barrier, while dysbiosis amplifies inflammation. - Restoring microbial balance and gut integrity is vital for healthy aging. Thanks for tuning in! "75 Gut-Healing Strategies & Biohacks" Follow Chloe on Instagram @synthesisofwellness Follow Chloe on TikTok @chloe_c_porter Visit synthesisofwellness.com --- Support this podcast: https://podcasters.spotify.com/pod/show/chloe-porter6/support
In today's episode, we dive into the interplay between the gut and immune system, focusing specifically on the critical role of immunoglobulins in mucosal immunity. Immunoglobulins, specialized glycoproteins synthesized by B cells, serve as the frontline defenders within the gastrointestinal tract, where they neutralize pathogens and maintain immune homeostasis. We also explore the mechanisms by which these antibodies fortify the intestinal barrier and aid in preventing dysbiosis. We finish by going through tools for supporting immunoglobulins. Topics: 1. Introduction - Overview of the gut-immune axis and focus on immunoglobulins. - How low levels of immunoglobulins can contribute to dysbiosis. 2. What Are Immunoglobulins? - Immunoglobulins (Igs) are glycoproteins produced by B cells. - They play a critical role in recognizing and neutralizing pathogens. - Antigen-binding sites for specific targeting. - Essential for immune homeostasis and pathogen elimination. 3. Immunoglobulins' Role in the Gut - Protect mucosal surfaces exposed to antigens. - B cells differentiate into plasma cells, and secrete immunoglobulins into the gut lumen. 4. Layers of the Intestinal Lining - Intestinal lumen is covered by a protective mucus layer. - Mucosa consists of epithelial cells, including mucus-secreting goblet cells. - Lamina propria beneath the epithelium is rich in immune cells. - Submucosa beneath mucosa. 5. The Function of the Mucus Layer - Acts as a physical barrier against pathogens. - Functions as a biochemical barrier by housing antimicrobial peptides and immunoglobulins. - Primarily composed of mucins secreted by goblet cells. - Goblet cells release mucins in response to stimuli, forming gel-like mucus. 6. Introduction to Secretory Immunoglobulin A (sIgA) - sIgA is the most abundant immunoglobulin in mucosal secretions. - Forms a protective barrier in the mucus layer. - Plays a critical role in immune exclusion by trapping and neutralizing pathogens. - Helps maintain non-inflammatory defense at mucosal surfaces. 7. Production of Secretory IgA - Plasma cells in the lamina propria secrete IgA. - IgA binds to receptors on epithelial cells and is transported across to the gut lumen. - sIgA to neutralize pathogens effectively within the mucus layer. 8. Functions of Secretory IgA in the Gut - Binds to antigens like bacterial walls, viruses, and toxins. - Traps antigens in the mucus to prevent adherence to epithelial cells. - Provides immune exclusion, reducing pathogen access to the gut lining. - Maintains mucosal barrier integrity to prevent inflammation. 9. Impact of Low sIgA Levels - Contributes to dysbiosis by allowing pathogenic bacteria to proliferate. - Intestinal hyperpermeability. - Increased risk of intestinal inflammation and infections. 10. Comparison with Other Immunoglobulins: IgG and IgM - IgG is most abundant in blood. - IgM is produced early in immune responses. - IgM can also be secreted into the gut lumen in IgA deficiency. 11. Immunoglobulin Supplements - IgG supplements. - Colostrum: IgG to help neutralize pathogens. - Supports gut barrier integrity. 12. Supporting Immunoglobulin Levels - Colostrum - Probiotics, prebiotics - Vitamin A - Zinc 13. Conclusion - Immunoglobulins, intestinal health, and immune balance. Thanks for tuning in! "75 Gut-Healing Strategies & Biohacks" Follow Chloe on Instagram @synthesisofwellness Follow Chloe on TikTok @chloe_c_porter Visit synthesisofwellness.com --- Support this podcast: https://podcasters.spotify.com/pod/show/chloe-porter6/support
In today's episode, we dive into the critical role of phospholipids in maintaining brain health, examining how these molecules contribute to neuronal communication, synaptic plasticity, and cognitive resilience. We'll explore the biochemical structure and function of key phospholipids like phosphatidylserine and phosphatidylcholine within neuronal membranes, where they play indispensable roles. We discuss associations and impacts of environmental toxins, aging, and neurodegenerative conditions. Finally, we discuss dietary tools to support phospholipid levels and promote cognitive health. Topics: 1. Introduction to Phospholipids and Cognitive Health 2. Brain Cell Structure and Composition - Neurons, glial cells. 3. Detailed Anatomy of Neurons - Soma contains organelles, dendrites receive signals, axon sends impulses. - Myelin sheath insulates axon, speeding signal transmission. - Myelin is rich in phospholipids. 4. Roles of Glial Cells in Brain Health - Astrocytes, oligodendrocytes, and microglia support neurons. - Glial cells regulate the brain's environment, form myelin, and provide immune defense. - Phospholipid-rich membranes are essential for glial function. 5. Biochemistry: Phospholipids - Phospholipids have a glycerol backbone, fatty acid tails, and a phosphate group. - Hydrophilic and hydrophobic parts form bilayers. - Key phospholipids: PC, PE, PS, PI 6. Phospholipid Bilayer's Role in Neuronal Communication - Ion channels, receptors, and transporters in the bilayer enable cell functions. - Ion channels allow ions to flow, creating signals for neuron communication. - Receptors detect neurotransmitters, initiating responses. 7. Neuronal Activation and Electrochemical Gradients - Resting neurons have ion concentration differences inside and outside the cell. - Ion channel activity during activation creates an action potential. - The phospholipid membrane enables controlled ion flow for signal transmission. 8. Neurotransmitter Release - Action potential at axon terminal triggers calcium entry. - Calcium causes vesicles to release neurotransmitters. - Released neurotransmitters bind to receptors, continuing the signal. 9. Diversity of Phospholipids in Neuronal Membranes - Different phospholipids (PC, PE, PS, PI) are essential for membrane integrity. 10. Summary: Phospholipids in Brain Function and Cognitive Health - Phospholipids support neuronal communication, synaptic plasticity, and cognitive resilience. - Synaptic plasticity - essential for learning and memory. 11. Phospholipid Disruption and Cognitive Decline - Oxidative stress, aging, and inflammation disrupt phospholipid composition. - Lipid peroxidation damages membranes, affecting neuron signaling. - Phospholipid damage contributes to cognitive decline. 12. Importance of Phospholipids in Aging and Brain Health - Lipid levels decrease with age, impacting brain function. 13. Environmental Toxins and the Brain - Heavy metals like mercury cause oxidative damage to phospholipids. - Damaged phospholipids and impaired neuron function. 14. Consequences of Suboptimal Phospholipids - Cognitive symptoms. - Low levels seen in neurodegenerative conditions. 15. Tools for Supporting Phospholipid Levels - Foods with PS and PC, such as fatty fish and eggs. - Phospholipid supplements. Thanks for tuning in! "75 Gut-Healing Strategies & Biohacks" Follow Chloe on Instagram @synthesisofwellness Follow Chloe on TikTok @chloe_c_porter Visit synthesisofwellness.com --- Support this podcast: https://podcasters.spotify.com/pod/show/chloe-porter6/support
In this episode, we explore serotonin synthesis within enterochromaffin (EC) cells in the gut, detailing how tryptophan is converted into serotonin through enzymatic processes. We examine the role of gut microbiota-derived metabolites, such as short-chain fatty acids (SCFAs), in modulating the synthesis of serotonin including impacting tryptophan hydroxylase activity. We explore serotonin's interactions with receptors on enteric neurons and vagal afferent fibers, analyzing how these signaling pathways influence gut motility. Finally, we uncover conditions and symptoms associated with low serotonin production and the importance of the intestinal microbiome. Topics: 1. Introduction to Gut-Produced Serotonin - Serotonin production within the gut. - Serotonin's role beyond mood. - Synthesis, causes of low serotonin, related GI symptoms. 2. Gut Lining Structure and Cell Types - Layers of the gut lining, focusing on the mucosa. - Description of epithelial cells, including enterocytes, goblet cells, Paneth cells, and enteroendocrine cells. - Role of enterochromaffin (EC) cells in serotonin production. 3. Serotonin Synthesis in EC Cells - Location and function of EC cells. - How EC cells synthesize serotonin from tryptophan. 4. Biochemical Pathway of Serotonin Production - Step-by-step process: conversion of tryptophan to serotonin. - Enzymes involved, including TPH1 and AADC. - Importance of tryptophan availability in serotonin synthesis. 5. Storage and Release of Serotonin in EC Cells - Role of VMAT1 in serotonin storage within vesicles. - Controlled release. 6. Triggers for Serotonin Release - Physical triggers: mechanical stretch, pressure from food intake. - Chemical triggers: microbial metabolites, bile acids. - Receptors involved (GPCRs, TGR5) and signaling pathways. 7. Release of Serotonin into Intestinal Lining Layers - Serotonin exocytosis and interaction with nearby cells. - Release of serotonin on both luminal and basolateral sides of EC cells. - How luminal and basolateral release affects gut motility and barrier function. 8. Serotonin's Role in Gut Motility - Interaction with 5-HT3 and 5-HT4 receptors on enterocytes and enteric neurons. - Activation of the enteric nervous system (ENS) in the submucosal and myenteric plexuses. - Coordination with pacemaker cells for peristaltic movement. 9. Immune Function and Serotonin in the Gut - Effect on immune cells. 10. Gut-Brain Communication via Serotonin and the Vagus Nerve - Activation of vagal afferent fibers by serotonin. 11. Contributing Factors to Low Serotonin Production - Impact of dysbiosis and reduced SCFA production. - SIBO specifically. - Intestinal inflammation in general. - Imbalanced microbiota and inflammation can disrupt EC cell function. 12. Manifestations of Low Serotonin in the Gut - Effects on motility: constipation, dysmotility... - Common GI symptoms, including bloating, discomfort, and fullness. - Association with conditions like IBS. 13. Supporting Serotonin Production in the Gut - Painting a full picture and identifying root causes. - Strategies to foster a healthy gut microbiome. - Role of sunlight and tryptophan-rich foods in serotonin production. - Stool testing for microbiome imbalances. Thanks for tuning in! "75 Gut-Healing Strategies & Biohacks" Follow Chloe on Instagram @synthesisofwellness Follow Chloe on TikTok @chloe_c_porter Visit synthesisofwellness.com --- Support this podcast: https://podcasters.spotify.com/pod/show/chloe-porter6/support
In this episode, we explore the role of Vitamin D in brain health, focusing on how its active form, calcitriol (1,25-dihydroxyvitamin D3), influences cognitive function and neuroprotection. We'll discuss the synthesis of Vitamin D, its conversion into its active form, and its ability to cross the blood-brain barrier to exert effects on neurons and glial cells. Additionally, we will highlight how calcitriol regulates gene expression through Vitamin D Response Elements (VDREs), impacting inflammation, antioxidant defense, and neurotrophic support. Finally, we touch on the symptoms of low Vitamin D and the link to cognitive decline. Topics: 1. Introduction to Vitamin D and the Brain - Forms of Vitamin D, synthesis, conversion/activation, and its role in the brain 2. Vitamin D - Vitamin D2 (ergocalciferol), plant-based sources like mushrooms - Vitamin D3 (cholecalciferol), UVB radiation, animal-based foods - Inactive forms 3. Synthesis and Conversion of Vitamin D3 - Synthesis in the skin: 7-dehydrocholesterol converts to pre-vitamin D3 - Conversion in the liver: 25-hydroxylase converts D3 to 25-hydroxyvitamin D (calcidiol) - Final activation in the kidneys: 1-alpha-hydroxylase converts calcidiol to 1,25-dihydroxycholecalciferol (calcitriol), the active form 4. Calcitriol and the Blood-Brain Barrier - Calcitriol's lipophilic nature, crossing the BBB - The structure and function of the blood-brain barrier - How calcitriol diffuses across the BBB and reaches brain cells 5. Calcitriol's Role in the Brain - Interaction with Vitamin D receptors (VDRs) in neurons and glial cells - VDRs in key brain regions: hippocampus, prefrontal cortex, cerebellum, basal ganglia - Binding of calcitriol to VDRs, conformational change, and formation of the VDR-RXR complex 6. Gene Regulation via Vitamin D Response Elements (VDREs) - Overview of VDREs in promoter regions of genes - Role of calcitriol in activating or repressing gene transcription 7. Impact on Inflammatory Responses - VDREs in anti-inflammatory genes promote IL-10 expression - Calcitriol's role in reducing pro-inflammatory cytokines like IL-6 and TNF-alpha 8. Neurotrophic Factors - VDREs' role in regulating genes that promote BDNF - BDNF's impact on neuron survival, growth, and synaptic connectivity 9. Antioxidant Enzymes - VDREs influence the expression of glutathione peroxidase and superoxide dismutase (SOD) - The role of these enzymes in defending neurons from oxidative stress 10. Brain Regions & Calcitriol - Hippocampus: Learning, memory, neurogenesis, synaptic plasticity - Prefrontal Cortex: Executive functions, mood regulation - Cerebellum: Motor control, cognitive processing, calcium homeostasis - Basal Ganglia: Movement regulation, protection of dopaminergic neurons - Amygdala: Emotion processing, fear, anxiety, stress response 11. Interconnection of Brain Regions - How Vitamin D's effects on neurotransmitter regulation, anti-inflammatory action, and calcium homeostasis create a global protective effect. 12. Conclusion - Recap of calcitriol's cellular mechanisms and neuroprotective effects - Symptoms of low Vitamin D Thank you to our episode sponsor: 1. Check out Daily Nouri and use code CHLOE20 for 20% off your order. Thanks for tuning in! Get Chloe's Book Today! "75 Gut-Healing Strategies & Biohacks" Follow Chloe on Instagram @synthesisofwellness Follow Chloe on TikTok @chloe_c_porter Visit synthesisofwellness.com to purchase products, subscribe to our mailing list, and more! --- Support this podcast: https://podcasters.spotify.com/pod/show/chloe-porter6/support
We are honored to be joined by Carri Norton and Ron Butterworth, the Co-Founders of the Sparkle Revolution App. Carri is an accomplished professional in software application development, with experience across various industries such as educational software, solar monitoring, energy management, and HVAC digital solutions. Starting her career as a software developer, she progressed into management and leadership roles, contributing to both startups and Fortune 500 companies. Her passion lies in creating user-centric software solutions that enhance customer experiences and make lives better. Ron's professional career has been that of a hardware design engineer. He has had the opportunity to work on projects based in education, solar energy, agriculture, and augmented reality, to name a few. Together they were thrilled to combine their expertise with personal passions and launch Sparkle Revolution. Topics: 1. Motivation to Create the App / Personal Journeys for Ron and Carri - Inspiration behind the app - Personal stories of Ron and Carri - Introduction to the app and its 4 "foundations" 2. How the App's Foundations Relate to Health - Overview of the 4 foundations - Impact of these foundations on mental, emotional, and physical health 3. Science and Studies on Mindfulness and Gratitude - Research on mindfulness practices - Scientific evidence on the benefits of gratitude 4. Examples of Mindfulness Exercises - Simple exercises for beginners - Techniques used to enhance daily mindfulness 5. Manifestation and Visualization Techniques - How manifestation works on a psychological level - Rewiring the brain to embrace future opportunities 6. Personal Examples of Manifestation in Action - Stories from Ron and Carri's lives - Real-world outcomes from practicing manifestation 7. Strategies for Integrating Mindfulness and Building Habits - Daily habits to foster mindfulness - Tips for sustainable habit formation 8. Overview of the Sparkle Revolution App - How the app works - Features and functionality - Where to download the app 9. Social Handles, Website, and Other Resources - Social media platforms - Official website and contact info - Additional tools or resources for further exploration To learn more and get the app, check out www.sparklerevolution.com Search for Sparkle Revolution on the Apple or Android stores. Visit them on social media @SparkleRevolutionApp Take advantage of an introductory offer! Use code CHLOE60 to get 60% off an annual subscription. Studies & Articles Mentioned: Robert Emmons, Why Gratitude is Good Article on National Library of Medicine about the benefits of mindfulness https://sparklerevolution.com/mindfulness-and-happiness/ --- Support this podcast: https://podcasters.spotify.com/pod/show/chloe-porter6/support
In this episode, we dive into the role of microglial cells, the brain's resident immune defenders, and how their chronic activation at the cellular level contributes to neurodegeneration. We'll explore the cellular mechanisms behind microglial activation, including the involvement of P2Y12 receptors and the release of pro-inflammatory cytokines like IL-1β and TNF-α. Additionally, we'll discuss how everyday environmental toxins and stressors can trigger long-term microglial activity, potentially increasing the risk of conditions like Alzheimer's. Topics: 1. Introduction - Overview: Microglia and neurodegeneration. - Environmental toxins affecting microglia. 2. Brain and Microglial Overview - Brain has neurons and glial cells. - Microglia are the brain's immune cells. - Roles: Immune defense, synaptic pruning, neuroprotection. 3. Microglial Immune Function - Constantly monitor for infection and damage. - Activated microglia perform phagocytosis. - Clear debris and maintain brain health. 4. Cellular Mechanisms of Activation - P2Y12 receptors respond to ATP/ADP. - Microglia shift from resting to activated. - Activated microglia are highly phagocytic. 5. Role in Synaptic Pruning - Microglia help in synaptic pruning. - Remove weak synapses for efficient brain circuits. 6. Microglia in Neurodegeneration - Chronic activation leads to inflammation. - Release of cytokines like IL-1β, TNF-α, IL-6. - Contributes to Alzheimer's, Parkinson's. - MHC molecules 7. Environmental Toxins and Activation - Mycotoxins cross BBB and activate microglia - Heavy metals like lead, aluminum affect neurons - Pesticides/herbicides linked to Parkinson's risk 8. Other Factors Activating Microglia - Industrial chemicals, BPA - Artificial additives, alcohol - Chronic stress 9. Conclusion - Recap: Microglial functions and overactivation. - Lifestyle factors influence microglial health. Thank you to our episode sponsors: 1. Check out Daily Nouri and use code CHLOE20 for 20% off your order. 2. Check out the TruAge Biological Age Test from TruMe Labs. Thanks for tuning in! Get Chloe's Book Today! "75 Gut-Healing Strategies & Biohacks" Follow Chloe on Instagram @synthesisofwellness Follow Chloe on TikTok @chloe_c_porter Visit synthesisofwellness.com to purchase products, subscribe to our mailing list, and more! --- Support this podcast: https://podcasters.spotify.com/pod/show/chloe-porter6/support
In this episode, we explore the critical role of Secretory IgA (sIgA) in maintaining intestinal health. We discuss how sIgA acts as a frontline immune defense, preventing pathogens from adhering to the gut epithelium and aiding in controlling microbial populations. Additionally, we dive into the implications of low and high sIgA levels - as indicated in a stool test, how sIgA levels relate to dysbiosis and Candida overgrowth, addressing underlying root causes, and tools for supporting healthy sIgA levels. Topics: 1. Introduction to Secretory IgA and its Functions - Overview of sIgA's role in mucosal immunity - Importance of sIgA in maintaining the gut's lining integrity - How sIgA regulates pathogenic microbes in the gut 2. Structure of the Gut Lining and Mucosal Layers - The intestinal lumen and the protective mucus layer - Layers of the intestinal wall: mucosa, submucosa, muscularis, and serosa - The epithelium within the mucosa: epithelial cells including goblet cells 3. The Mucus Layer and Its Protective Function - Composition of the mucus layer: mucins and other components - How the mucus acts as both a physical and biochemical barrier - Differences between the mucus layer in the small and large intestines 4. Secretory IgA: Location and Role in the Gut - Where sIgA is found in the mucus layer - Differences in sIgA concentration in the small vs. large intestine - Interaction between sIgA and mucins to trap pathogens 5. Production and Secretion of Secretory IgA - Plasma cells in the lamina propria and their role in sIgA production - Transport of sIgA across the gut epithelium via the polymeric immunoglobulin receptor (pIgR) - Secretory component (SC) and its role in protecting sIgA from degradation 6. Immune Functions of sIgA in the Gut - Binding and neutralizing pathogens - The process of immune exclusion - The role of sIgA in preventing pathogen adherence to epithelial cells 7. Low and High sIgA Levels and Gut Conditions - What low and high sIgA levels can indicate in a stool test - Gut conditions associated with abnormal sIgA levels (e.g., dysbiosis, Candida overgrowth, infections) - The impact of chronic stress and prolonged infections on sIgA levels 8. Root Causes for Low sIgA Levels - Addressing root causes and underlying infections - Lifestyle factors that support healthy sIgA levels - Nutrients such as Vitamin A and Zinc for supporting sIgA production 9. Probiotics & More to Support sIgA - Probiotics, prebiotics, and phages that enhance sIgA production - Individual gut health needs Thank you to our episode sponsors: 1. Check out Daily Nouri and use code CHLOE20 for 20% off your order. 2. Check out the TruAge Biological Age Test from TruMe Labs. Thanks for tuning in! Get Chloe's Book Today! "75 Gut-Healing Strategies & Biohacks" Follow Chloe on Instagram @synthesisofwellness Follow Chloe on TikTok @chloe_c_porter Visit synthesisofwellness.com to purchase products, subscribe to our mailing list, and more! --- Support this podcast: https://podcasters.spotify.com/pod/show/chloe-porter6/support
In this episode, we explore the critical roles of the enteric nervous system (ENS) and the vagus nerve in digestion and gut motility. We also examine how autonomic nervous system dysfunction can contribute to conditions such as Small Intestinal Bacterial Overgrowth (SIBO). Topics: 1. Introduction to the Nervous System and Its Role in Digestion - Overview of the enteric nervous system (ENS) and vagus nerve - Focus on location, function, and associated dysfunctions 2. Overview of the Enteric Nervous System (ENS) - ENS as one of the three divisions (Sympathetic, Parasympathetic, Enteric) - Location and function overview 3. The Mucosa - Structure and Function of the Mucosa - Epithelium (cells such as enterocytes, goblet cells, enteroendocrine cells) - Lamina propria (loose connective tissue, immune cells) - Smooth muscle layer 4. The Submucosa - Structure and Function of the Submucosa - Thicker connective tissue providing support for blood vessels, lymphatics, and nerves 5. The Submucosal Plexus (Part of the ENS) - Location and Functions of the Submucosal Plexus - Regulation of digestive enzyme secretion, mucin production, and electrolyte balance - Role in creating protective mucus barriers and controlling local blood flow 6. Mucus Secretion and Protection - Role of Goblet Cells - Secretion of mucins and formation of the protective mucus layer 7. Blood Flow Regulation by the Submucosal Plexus - Role in adjusting blood supply during digestion 8. Neuronal Cells of the Submucosal Plexus - Sensory Neurons: Detecting changes in the gut - Interneurons: Processing and integrating sensory input - Secretomotor Neurons 9. Communication Between Neurons and Epithelial Cells - Chemical Signaling and Neurotransmitters - ACh and Vasoactive Intestinal Peptide (VIP) - Serotonin's role in activating sensory neurons and regulating secretion 10. Role of the Vagus Nerve in Digestive Regulation - Vagus Nerve as Part of the ANS - Regulation of digestive secretions, gut motility, and smooth muscle contractions 11. Small Intestinal Bacterial Overgrowth (SIBO) - Connection Between Autonomic Nervous System Dysfunction and SIBO - Reduced motility leading to bacterial overgrowth - Impact on gut environment - Root cause approach Thanks for tuning in! Get Chloe's Book Today! "75 Gut-Healing Strategies & Biohacks" Follow Chloe on Instagram @synthesisofwellness Follow Chloe on TikTok @chloe_c_porter Visit synthesisofwellness.com to purchase products, subscribe to our mailing list, and more! --- Support this podcast: https://podcasters.spotify.com/pod/show/chloe-porter6/support
We are honored to be joined by Dr. Diana Driscoll. An authority on the autonomic nervous system, Dr. Diana Driscoll, Optometrist, FAAO, is a world-renowned expert on POTS (Postural Orthostatic Tachycardia Syndrome), and other “invisible” illnesses. As an author, speaker, inventor, and devoted researcher with five patents to date, she continues to push forward with dramatic shifts in the evaluation and treatment of these conditions. Dr. Driscoll's personal battle with POTS inspired a decade of self-funded groundbreaking research and innovative treatment. Now an authority on POTS, IIH, ME/CFS, the vagus nerve, dry eye disease, neurological gastroparesis, and brain health, she offers the compassion of a former patient, as well as strong science. Topics: 1. Introduction and Personal Journey - Discussion of Dr. Diana Driscoll's personal battle with POTS and how it led to her groundbreaking research. 2. Overview of POTS (Postural Orthostatic Tachycardia Syndrome) - Diagnostic criteria and common symptoms. - How POTS develops. - The body systems affected by POTS and how they are impacted. 3. Inflammatory POTS - Definition and introduction to "Inflammatory POTS" 4. Nervous System and the Vagus Nerve, Specifically - Further exploration of the role of the nervous system in POTS. - The impact of the vagus nerve on various body functions, including digestion and bowel movements. - Understanding chronic dry eyes. 5. Supplementation and Solutions - TJ Nutrition resources and supplements. - Parasym Plus™ to support the vagus nerve and acetylcholine functions. - Beta Balance™ NAC MAX™ Check out TJ Nutrition and Dr. Diana Driscoll's supplements here and use code PORTER10 to get 10% off. Thanks for tuning in! Get Chloe's Book Today! "75 Gut-Healing Strategies & Biohacks" Follow Chloe on Instagram @synthesisofwellness Follow Chloe on TikTok @chloe_c_porter Visit synthesisofwellness.com to purchase products, subscribe to our mailing list, and more! --- Support this podcast: https://podcasters.spotify.com/pod/show/chloe-porter6/support
In today's episode, we dive into the molecular mechanisms underlying neuroinflammation, with a particular focus on how gut-derived endotoxins, such as lipopolysaccharides (LPS), perturb the blood-brain barrier (BBB) and propagate neuroinflammatory cascades. In more detail, we'll discuss how intestinal dysbiosis and increased intestinal permeability can allow endotoxins such as LPS to enter systemic circulation and cross the blood-brain barrier (BBB), where they activate microglia via the TLR4 signaling pathway. We will explore symptoms including brain fog, memory impairment, mood disturbances, decreased concentration, and cognitive fatigue; as well as tools to support the gut-microbiota-brain axis. Topics: 1. Introduction to Neuroinflammation - Definition and general overview - Key brain cells: neurons, glial cells, and endothelial cells - Chronic neuroinflammation and microglial cells 2. Cellular Mechanisms of Neuroinflammation - Role of microglia in detecting damage or infection - Microglial activation through PRRs/TLRs - Release of pro-inflammatory cytokines and reactive oxygen species (ROS) - Impact on neurons and synaptic plasticity: memory and learning 3. Astrocytes in Neuroinflammation - Astrocytes' contribution to the inflammatory response 4. Chronic Neuroinflammation and Brain Health - Prolonged activation: oxidative stress, excitotoxicity, impaired synaptic function - Impairment of synaptic plasticity and cognitive decline - Mitochondrial dysfunction and cell death cascades - Compromised blood-brain barrier integrity 5. Contributing Factors to Neuroinflammation - Environmental toxins and pollutants, viral or bacterial infections, chronic sleep deprivation, and more 6. Intestinal Dysbiosis and Neuroinflammation - Role of Gram-negative bacteria and LPS (lipopolysaccharides) - Increased intestinal permeability and passage of LPS - LPS transport: transcellular and paracellular pathways - Impact on the blood-brain barrier (BBB) 7. LPS Activation of Microglia - LPS binding to TLR4 on immune cells - Microglial activation and cytokine release - Cognitive impairments and neurodegenerative processes 8. Intestinal Hyperpermeability - Bioindividual approaches: prebiotics, probiotics, polyphenols, bacteriophages - Support for intestinal mucus secretions and sources of mucilage - Butyrate - Glutamine 9. Beneficial Gut Microbiome-Derived Metabolites and Brain Health - Types of metabolites: SCFAs, tryptophan metabolites, polyphenol metabolites - Role of butyrate in crossing the BBB and modulating brain function - Influence on gene expression and synaptic plasticity - Contribution to gut barrier integrity and protection against endotoxins 10. Conclusion - Recap of neuroinflammation's impact on cognitive function + symptoms Thank you to our episode sponsor: 1. Check out Daily Nouri and use code CHLOE20 for 20% off your order. Thanks for tuning in! Get Chloe's Book Today! "75 Gut-Healing Strategies & Biohacks" Follow Chloe on Instagram @synthesisofwellness Follow Chloe on TikTok @chloe_c_porter Visit synthesisofwellness.com to purchase products, subscribe to our mailing list, and more! --- Support this podcast: https://podcasters.spotify.com/pod/show/chloe-porter6/support
In this episode, we dive into how short-chain fatty acids (SCFAs), particularly butyrate, and tryptophan metabolites support the gut-immune axis by promoting regulatory T cell differentiation, strengthening the intestinal barrier, and enhancing antimicrobial defenses through IL-22 signaling. We break down how butyrate can improve immune tolerance as well as epithelial integrity, aiding in the prevention of chronic inflammatory responses. We also detail practical ways to support butyrate levels and aid in strengthening both the epithelial barrier and gut-immune axis. Topics: 1. Introduction - Overview of the role of SCFAs and tryptophan metabolites in supporting the gut-immune axis. - Quick review of the location of immune cells in relation to the gut microbiota. 2. The Intestinal Barrier - Structure of the intestinal wall and layers - Focus on the mucosal layer, specifically epithelium and lamina propria. 3. The Lamina Propria - Structural elements: fibroblasts, extracellular matrix (ECM), and myofibroblasts. - Vascular components: endothelial cells, capillaries, and lymphatic vessels. - Importance of the lamina propria as a hub for immune responses. 4. Immune Cells in the Lamina Propria - T cells: Role of regulatory T cells (Tregs) in immune modulation. - B cells: Production of IgA, class switching, and plasma cells. - Dendritic cells: Sampling luminal antigens and initiating immune responses. - Macrophages: Phagocytic activity, pro-inflammatory (M1) vs. anti-inflammatory (M2) states. - Mast cells: Role in allergic responses, chronic inflammatory conditions, and MCAS. 5. Short-Chain Fatty Acids (SCFAs) - Production of SCFAs (acetate, propionate, butyrate) by gut microbiota. - Butyrate's role in supporting regulatory T cell (Treg) differentiation and immune tolerance. -Butyrate as fuel for epithelial cells and the production of tight junction proteins. 6. Mechanisms of Butyrate in Immune Modulation - Impact on Tregs through FoxP3 expression. - SCFA's role in maintaining immune balance. 7. Butyrate and Epithelial Integrity - Support for tight junction protein expression. - Prevention of translocation of harmful microbes and antigens. - Reduced systemic inflammation through a strengthened barrier. 8. Supporting Butyrate Production - Sodium butyrate supplementation and microbiome optimization. - Role of fiber, polyphenols, and probiotics. 9. Tryptophan Metabolites - Overview of tryptophan metabolism by gut bacteria into indoles. - Indoles' role in promoting IL-22 production, contributing to antimicrobial defense and immune tolerance. 10. IL-22 - IL-22's enhancement of antimicrobial peptides (AMPs) and mucin production. - Case Study: Role of Lactobacillus strains in restoring IL-22 and helping to mitigate colitis. 11. Conclusion - Recap of how SCFAs and tryptophan metabolites interact with the gut-immune axis. - Importance of gut microbiome support for maintaining immune balance. Thank you to our episode sponsor: 1. Check out Daily Nouri and use code CHLOE20 for 20% off your order. Thanks for tuning in! Get Chloe's Book Today! "75 Gut-Healing Strategies & Biohacks" Follow Chloe on Instagram @synthesisofwellness Follow Chloe on TikTok @chloe_c_porter Visit synthesisofwellness.com to purchase products, subscribe to our mailing list, and more! --- Support this podcast: https://podcasters.spotify.com/pod/show/chloe-porter6/support
In this episode, we explore the crucial role of the protective mucus layer in maintaining intestinal lining integrity, highlighting how it shields the epithelial cells from harmful substances and supports overall digestive function. We dive into the components and functions of mucins, goblet cells, and secretory IgA, explaining their contributions to gut immunity and barrier integrity. Additionally, we discuss strategies for supporting mucus production centered around butyrate, N-acetyl glucosamine, and plant-derived mucilage. Topics: 1. Introduction: Overview of the Gastrointestinal Mucus Layer - Importance of the mucus secretions in forming a protective layer - Brief review of the gastrointestinal lining and its structure 2. The Mucosal Layer and Its Components - The mucus layer on the epithelial cells of the mucosa - Four main layers of the intestinal wall, focusing on the mucosa - Epithelium: absorptive enterocytes and mucus-secreting goblet cells - Lamina propria: connective tissue rich in immune cells, blood vessels, and lymphatics - Smooth muscle aiding in subtle movements - Submucosa: thicker connective tissue layer with blood vessels, nerves, and lymphatics 3. Dual Function of the Mucus Layer - Physical barrier preventing pathogens and toxins from reaching epithelial cells - Biochemical barrier housing antimicrobial peptides and immunoglobulins 4. Goblet Cells and Their Role in Mucin Production - Goblet cells: specialized epithelial cells producing mucins - Distribution of goblet cells from small to large intestine - Importance of goblet cells in maintaining the mucus barrier 5. Mucins: Structure and Glycosylation - Glycosylation and its role in mucus properties and host-microbiota interactions - Commensal bacteria and their interactions with mucins (e.g., Bacteroides and Akkermansia muciniphila) 6. The Importance of Controlled Mucin Degradation - Mutualistic relationship between mucin-degrading bacteria and the host - Risks of dysregulated mucin degradation, leading to dysbiosis and leaky gut 7. Secretory IgA and Its Role in Gut Immunity - Overview of secretory IgA (sIgA) and its production - sIgA's mechanism of neutralizing pathogens "without inflammation" - Immune exclusion as a defense mechanism to maintain gut barrier integrity 8. Recap: The Dynamic Nature of the Mucus Layer - Continuous renewal of the mucus layer and the role of goblet cells and plasma cells - Importance of regular mucus turnover for maintaining a healthy gut barrier 9. Supporting Gastrointestinal Mucus Production - Short-chain fatty acids (SCFAs) and their role in stimulating mucus production - Butyrate and its effects on goblet cells and epithelial repair - N-Acetyl Glucosamine (NAG) as a supplement to support mucin glycosylation 10. Supporting the Intestinal Lining After Dysbiosis or Increased Permeability - Mucilage and its role in coating and soothing mucosal surfaces - Plant-derived mucilage from slippery elm and marshmallow root - Physical protection provided by mucilage - Soothing effects on inflamed tissues and promoting smooth digestion Thank you to our episode sponsor: 1. Check out Daily Nouri and use code CHLOE20 for 20% off your order. Thanks for tuning in! Get Chloe's Book Today! "75 Gut-Healing Strategies & Biohacks" Follow Chloe on Instagram @synthesisofwellness Follow Chloe on TikTok @chloe_c_porter Visit synthesisofwellness.com to purchase products, subscribe to our mailing list, and more! --- Support this podcast: https://podcasters.spotify.com/pod/show/chloe-porter6/support
In this episode, we explore the neuroanatomical and molecular mechanisms underlying sleep regulation, focusing on the interplay between the suprachiasmatic nucleus, brain stem, and pineal gland in maintaining circadian rhythms. We dive into the biochemical pathways involving neurotransmitters such as GABA and hormones like melatonin, elucidating their synthesis and role in sleep-wake transitions. Further, the discussion extends to practical strategies, emphasizing the influence of light exposure on melatonin production and the potential benefits of specific amino acid supplementation in supporting sleep quality. Additionally, we address the impact of chronic inflammation and other root causes of sleep disturbances, exploring how systemic health issues can disrupt sleep. Topics: 1. Introduction to Sleep Neuroanatomy - Overview of key brain structures involved in sleep regulation - Emphasis on the hypothalamus, brain stem, thalamus, basal forebrain, and pineal gland 2. Deep Dive into Key Brain Structures - Hypothalamus - Detailed role of the suprachiasmatic nucleus (SCN) in circadian rhythm regulation - Connection between SCN, light perception, and neuronal control - Brain Stem - Description of sleep-wake transitions mediated by the midbrain, pons, and medulla - Mechanisms of GABA production and its impact on inhibitory neurotransmission - Thalamus - Functions as a sensory relay - Pineal Gland - Regulation of melatonin synthesis, influenced by light exposure and SCN signaling - Basal Forebrain - Neuronal mechanisms promoting alertness and facilitating sleep onset - Role of adenosine accumulation in sleep pressure and its neurochemical effects 3. Mechanisms of Sleep Regulation - Exploration of the circadian rhythm's molecular underpinnings involving CLOCK and BMAL1 genes - Feedback loops involving period and cryptochrome proteins, detailing their synthesis and degradation within the SCN - Hormonal regulation by the SCN through cortisol and melatonin 4. Strategies to Support Sleep Quality - Importance of synchronizing light exposure with natural environmental cycles - Supplementation strategies based on literature: - GABA and L-theanine supplementation supporting inhibitory signaling - L-tryptophan may support serotonin and melatonin synthesis 5. Addressing Underlying Root Causes and Sources of Chronic Inflammation Contributing to Poor Sleep - Role of neuroinflammation and gut dysbiosis in sleep disruption - Discussion on neurotransmitters synthesized in the gut - Hormonal impacts Thank you to our episode sponsor: 1. Check out Liver Medic and use code Chloe20 to save 20% on "Leaky Gut Repair" Brendan's YouTube Channel https://x.com/livermedic Thanks for tuning in! Get Chloe's Book Today! "75 Gut-Healing Strategies & Biohacks" Follow Chloe on Instagram @synthesisofwellness Follow Chloe on TikTok @chloe_c_porter Visit synthesisofwellness.com to purchase products, subscribe to our mailing list, and more! --- Support this podcast: https://podcasters.spotify.com/pod/show/chloe-porter6/support
In today's episode, we explore the complex interactions between Candida and the gastrointestinal tract, examining how this fungus can transition from harmless commensal to pathogenic state that disrupts gut barrier functions. We dive into Candida's ability to undermine mucosal integrity through the production of candidalysin and other virulence factors like secreted aspartyl proteases, which specifically target and degrade tight junction proteins such as claudins and occludins. We'll explore the layered structure of the intestinal mucus barrier, its role in immunological defense, and how Candida's biofilm-forming capabilities further facilitate its pathogenicity. Lastly, we'll discuss tools and strategies for mitigating Candida overgrowth. Topics: 1. Gastrointestinal Tract Anatomy / Barrier Function - Mucus Layer - Description of the gastroprotective mucus barrier - Differences in mucus architecture between the small and large intestines - Composition: mucins, immunoglobulins (IgA), antimicrobial peptides - Intestinal Epithelium - Structure and function of the epithelial cell layer - Roles of different epithelial cells: enterocytes, goblet cells, enteroendocrine cells, Paneth cells - Lamina Propria - Immune cell types 2. Integrity of Tight Junctions - Tight junction proteins: claudins, occludins, junctional adhesion molecules (JAMs) - Mechanisms of tight junction regulation and response to changes in the gut environment - Impact of physiological and pathological factors on tight junction dynamics 3. Candida Pathogenicity Mechanisms - Adherence and Biofilm Formation - Mechanisms facilitating Candida adherence to epithelial cells - Structural and functional implications of biofilm formation - Enzymatic Disruption of Tight Junctions - Role of Candida-secreted aspartyl proteases (Sap) and phospholipases in degrading tight junction components - Immune System Interaction - Triggering of inflammatory cytokines by Candida presence and their effects on gut barrier function 4. Candidalysin: A Specific Virulence Factor - Genetic basis for candidalysin production via the ECE1 gene - Mechanism of action: disruption of epithelial cell membrane integrity, pore formation, and cellular damage 5. Clinical Manifestations of Candida Overgrowth - Gastrointestinal symptoms: bloating, gas, diarrhea, constipation, abdominal pain - Systemic effects: fatigue, brain fog, skin issues, mood disturbances - Nutrient absorption impairments leading to deficiencies 6. Strategies - Biofilm Disruption - Agents targeting biofilm integrity and adherence properties - Antifungal and Enzymatic Tools - Natural antifungals - Role of enzymatic supplements in biofilm and cell wall degradation - Immune Modulation and Gut Microbiota Support - Immune-modulating agents - Probiotics and prebiotics to restore and maintain healthy gut flora Thank you to our episode sponsors: 1. Check out Daily Nouri and use code CHLOE20 for 20% off your order. 2. Check out Liver Medic and use code Chloe20 to save 20% on "Leaky Gut Repair" Brendan's YouTube Channel https://x.com/livermedic Thanks for tuning in! Get Chloe's Book Today! "75 Gut-Healing Strategies & Biohacks" Follow Chloe on Instagram @synthesisofwellness Follow Chloe on TikTok @chloe_c_porter Visit synthesisofwellness.com to purchase products, subscribe to our mailing list, and more! --- Support this podcast: https://podcasters.spotify.com/pod/show/chloe-porter6/support
In today's episode, we dive into the crucial roles that gut-derived microbial metabolites, particularly short-chain fatty acids and tryptophan metabolites, play in supporting the health of neurons and glial cells, ultimately promoting cognitive function. We explore how enhancing the body's natural production of these metabolites through a healthy gut microbiome and how strategic supplementation can both benefit brain health. The discussion includes detailed insights into the cellular mechanisms in the brain and practical strategies for fostering a healthy gut microbiome capable of producing high quantities of these beneficial compounds. Topics: 1. Introduction to Brain Cellular Makeup - Overview of Neurons and Glial Cells - Neurons: Structure and Function - Glial Cells: Types and Roles 2. Neuronal Communication and Synaptic Function - Neuronal Structure: Soma, Dendrites, Axon - Synaptic Communication: Presynaptic Terminal, Synaptic Cleft, Postsynaptic Membrane 3. Energy Demands - ATP Utilization and Energy Demands - Mitochondrial Function and Neuronal Vulnerability 4. Astrocytes and Microglial Cells - Astrocytes: Functions and Role in CNS Homeostasis - Microglial Cells: Immune Functions and Role in Neuroinflammation 5. Roles of Gut-Derived Microbial Metabolites in Supporting Brain Health - Overview of Gut-Derived Metabolites - Short-Chain Fatty Acids (SCFAs): Acetate, Propionate, Butyrate - Tryptophan Metabolites: Indole, Indole-3-Propionate (IPA) 6. Impact of Gut-Derived Metabolites on Neurons - Promotion of Neurogenesis by SCFAs - Neuroprotective Effects of Tryptophan Metabolites 7. Impact of Gut-Derived Metabolites on Glial Cells - Impact on Astrocytes - Impact on Microglial Cells 8. Strategies to Enhance Metabolite Production - Diversity - Probiotic Supplementation - Fiber - Amino Acids 9. Direct Supplementation Options - Sodium Butyrate: Role in Neuroprotection and Cognitive Function Thank you to our episode sponsors: 1. Check out Daily Nouri and use code CHLOE20 for 20% off your order. 2. Check out AX3 Astaxanthin and use discount code CHLOE20 for 20% off your first order. 3. Check out Liver Medic and use code Chloe20 to save 20% on "Leaky Gut Repair" Brendan's YouTube Channel https://x.com/livermedic Thanks for tuning in! Get Chloe's Book Today! "75 Gut-Healing Strategies & Biohacks" Follow Chloe on Instagram @synthesisofwellness Follow Chloe on TikTok @chloe_c_porter Visit synthesisofwellness.com to purchase products, subscribe to our mailing list, and more! --- Support this podcast: https://podcasters.spotify.com/pod/show/chloe-porter6/support
In this episode, we delve into the complexities of how Borrelia burgdorferi, the bacterium responsible for Lyme disease, impacts the brain, leading to neuroinflammation and a spectrum of neurological symptoms. We'll explore the pathophysiology of Borrelia's interaction with the brain's blood-brain barrier, its evasion of the immune system, and the direct effects on neural cells. Additionally, we'll discuss specific neurological symptoms and manifestations. Topics: 1. Introduction - Overview of Lyme disease and its causative agent, Borrelia burgdorferi. - Explanation of Borrelia's invasive nature and its ability to cross biological barriers like the blood-brain barrier (BBB). 2. Understanding the Blood-Brain Barrier (BBB) - Endothelial Cells: Primary cellular component of the BBB, unique to the brain for their tightly joined structure. - Astrocytes: - Role in regulating blood flow through neurovascular coupling. - Interaction with neurons and release of vasoactive substances. - Contribution to BBB integrity by influencing tight junction formation. - Pericytes: - Contribution to BBB permeability and regulation through contractile capabilities. 3. Sponsor Break: Daily Nouri 4. Pathophysiology of Borrelia in the Brain - Invasion Mechanisms: - Borrelia's ability to adhere to and invade endothelial cells. - Role of surface proteins and induction of matrix metalloproteinases (MMPs). - Immune Evasion and CNS Invasion: - Antigenic variation to avoid immune detection. - Triggering of immune responses within the CNS, involving microglia and peripheral immune cells. - Cytokine production and its consequences on neuronal health. 5. Direct Impact on Brain Cells - Neurons: Effects of Borrelia invasion on cellular functions, mitochondrial dysfunction, and apoptosis. - Astrocytes: Transition to reactive astrocytes in the presence of Borrelia and their role in neuroinflammation. - Oligodendrocytes: Impact on myelin production and implications for cognitive and motor functions. 6. Sponsor Break: AX3 Life 7. Neurological Symptoms and Manifestations - Symptoms Overview: Headaches, facial palsy, cognitive impairments, nerve root inflammation, POTS. - Psychiatric Manifestations: Potential psychological symptoms like depression and anxiety due to neuroinflammation. 8. Conclusion - Importance of addressing neurological symptoms of Lyme through a bioindividual and root-cause approach. - Recap of key points and reference to previous relevant episodes for further listening. Thank you to our episode sponsors: Check out Daily Nouri and use code CHLOE20 for 20% off your order. Check out AX3 Astaxanthin and use discount code CHLOE20 for 20% off your first order. Thanks for tuning in! Get Chloe's Book Today! "75 Gut-Healing Strategies & Biohacks" Follow Chloe on Instagram @synthesisofwellness Follow Chloe on TikTok @chloe_c_porter Visit synthesisofwellness.com to purchase products, subscribe to our mailing list, and more! --- Support this podcast: https://podcasters.spotify.com/pod/show/chloe-porter6/support
We are so honored to be joined by Dave Watumull. Dave has more than twenty years of experience in the development and commercialization of astaxanthin—a safe and powerful, naturally-occurring molecule with important health and longevity benefits. He is the Co-founder and CEO of AX3 Life, the consumer health company behind the breakthrough product, AX3® Bio-Pure Astaxanthin. He is also the COO of Cardax, a life sciences business focused on advancing pharmaceutical and nutraceutical applications of astaxanthin. Topics: 1. Introduction to Astaxanthin - Background on its history and role in nature. - Ex: salmon vitality and color. 2. Mechanism of Action - How it localizes in cellular membranes. - Neutralizes free radicals and oxidative stress. - Fights inflammation to support whole body health and longevity. 3. Safety Profile - Exceptional safety supported by human clinical studies and rigorous toxicity testing. - Differentiation from other antioxidants, anti-inflammatories, and anti-aging products. 4. Health Benefits - Joint health - Topicals for skin - Cardiovascular health - Crossing the blood-brain barrier 5. Longevity Research - Discussion of longevity research with astaxanthin across multiple species. - Introduction to the NIH longevity study with AX3 Astaxanthin. 6. NIH Longevity Study Details - Overview of the NIH-funded Interventions Testing Program (ITP). - Discussion of the program's history, structure, and other notable agents tested. - Study results published with AX3 Astaxanthin. 7. Superior Features of AX3 Astaxanthin - Purity. - Description of the production process. - Human bioavailability data and environmental considerations. - The story behind "AX3" - Formulation insights Check out AX3 Astaxanthin Use discount code CHLOE20 for 20% off your order. Follow @ax3.life on Instagram. Thanks for tuning in! Get Chloe's Book Today! "75 Gut-Healing Strategies & Biohacks" Follow Chloe on Instagram @synthesisofwellness Follow Chloe on TikTok @chloe_c_porter Visit synthesisofwellness.com to purchase products, subscribe to our mailing list, and more! --- Support this podcast: https://podcasters.spotify.com/pod/show/chloe-porter6/support
In this episode, we dive into the incredible role of T4D-Myoviridae bacteriophages in targeting Escherichia coli strains, highlighting their unique contractile tail structures that facilitate precise DNA injection into host cells. We explore the specific interactions between phage tail fibers and bacterial lipopolysaccharide receptors, illustrating the phage's lock-and-key mechanism crucial for selective bacterial lysis. This discussion extends into the implications for combatting SIBO and enhancing gut microbiota balance while exploring the impressive scientific literature on phage-mediated modulation of gut flora and inflammatory markers. Topics: 1. Introduction to Bacteriophages - Explanation of what a bacteriophage (phage) is. - Importance of phages in research and their role in intestinal dysbiosis. 2. Bacteriophage Foundation - Description of bacteriophage structure: - Capsid and its genetic material (DNA or RNA). - Icosahedral head and its efficiency. - Types of phage tails and their functions: - Contractile tails: mechanism similar to a syringe. - Non-contractile tails: static structure for attachment. 3. Phage-Bacteria Interaction - Initial attachment to bacterial cell via tail fibers. - Specificity of attachment (lock and key mechanism). - Injection of genetic material into the bacterial cell. - Use of bacterial machinery to replicate phage components. 4. Phage Replication Cycle - Overview of the phage replication process within the host cell: - Integration and takeover of host cellular machinery. - Assembly of new phage particles. - Release of phages through lysis of the host cell. 5. Application - Potential of phages to selectively target and kill bacteria. - Benefits over broad-spectrum targeting. - Discussion of antibiotic resistance. 6. Diving into the Gut Microbiome - Connection between phages and the gut microbiome. - Discussion of specific phages like T4D-Myoviridae and their targets. - Relevance to SIBO (Small Intestinal Bacterial Overgrowth). 7. Case Studies and Research - Review of studies on phage impact on gut microbiota and inflammation. - Specific changes in microbiota observed in research (e.g., increase in butyrate-producing genera). Thank you to our episode sponsor: Liver Medic Use code Chloe20 to save 20% on "Leaky Gut Repair" Brendan's YouTube Channel https://x.com/livermedic Thanks for tuning in! Get Chloe's Book Today! "75 Gut-Healing Strategies & Biohacks" Follow Chloe on Instagram @synthesisofwellness Follow Chloe on TikTok @chloe_c_porter Visit synthesisofwellness.com to purchase products, subscribe to our mailing list, and more! --- Support this podcast: https://podcasters.spotify.com/pod/show/chloe-porter6/support
In this episode, we dive into the critical role of HLA gene variants, such as HLA-DRB1, and their profound impact on immune system function, particularly focusing on their significance in conditions like Lyme disease and mold toxicity. We'll explore how variations in these HLA genes, like the HLA-DRB1*0401 allele, influence the body's ability to recognize and respond to pathogens, potentially leading to chronic symptoms. The discussion will also cover HLA gene variant testing and how it can lend insight when it comes to taking a bioindividual approach to supporting the immune system. Topics: 1. Introduction - Explanation of HLA gene variants - Importance of HLA genes in immune system function 2. Basics of Genetics and Immunology - What is a chromosome? - Introduction to genes and their functions - Overview of DNA and protein synthesis 3. Major Histocompatibility Complex (MHC) - Description of the Major Histocompatibility Complex - Division of MHC genes into Class I, II, and III - Role of Class I MHC genes in antigen presentation - Role of Class II MHC genes in antigen presentation - Function of Class III MHC genes 4. HLA Genes and Immune Function - Explanation of Human Leukocyte Antigen (HLA) genes - Connection between MHC genes and HLA genes (human MHC) - Transcription and regulation of HLA genes - Response to internal and external signals - Role of Antigen-presenting cells (APCs) 5. Pathogen Recognition and Immune Activation - Mechanism of pathogen recognition by HLA genes - Activation of immune response by antigen presentation - Formation and function of peptide-HLA complexes 6. Impact of HLA Variants on Disease - Specific focus on Lyme disease and mold toxicity - Role of HLA Class II alleles in immune response - Association of HLA-DR alleles with chronic Lyme disease - Sensitivity to mold exposure linked to certain HLA gene variants 7. Testing and Implications of HLA Gene Variants - Importance of HLA gene variant testing - Broader implications for diseases like lupus and multiple sclerosis - Personalized approaches based on HLA gene variants 8. Biotoxin Illness and Poor Antigen Presentation Due to an HLA Gene Variant - Strategies to support immune system function in the presence of biotoxins - Importance of detoxification pathways and binders Thank you to our episode sponsor: Liver Medic Use code Chloe20 to save 20% on "Leaky Gut Repair" Brendan's YouTube Channel https://x.com/livermedic Thanks for tuning in! Get Chloe's Book Today! "75 Gut-Healing Strategies & Biohacks" Follow Chloe on Instagram @synthesisofwellness Follow Chloe on TikTok @chloe_c_porter Visit synthesisofwellness.com to purchase products, subscribe to our mailing list, and more! --- Support this podcast: https://podcasters.spotify.com/pod/show/chloe-porter6/support