Are Childhood Traumas Stored in the Cells of the Body?

SeljaSelja
18 min read

Introduction

Childhood trauma is no longer seen solely as a psychological or emotional injury; it is increasingly understood as a complex biological event that leaves lasting imprints within the body’s cells. These imprints influence gene expression, immune function, and cellular aging, thereby affecting an individual’s physical and mental health throughout life. The concept that trauma is “stored” in the cells challenges traditional notions of memory and healing, suggesting that trauma is a whole-body experience. This article explores the scientific evidence and mechanisms behind the cellular storage of childhood trauma, highlighting its implications for health and recovery.

2. Defining Childhood Trauma

Childhood trauma encompasses a wide range of adverse experiences, including physical, emotional, and sexual abuse, neglect, loss of caregivers, and chronic exposure to stress or violence. These events occur during critical developmental periods when the brain and body are highly plastic and vulnerable. Trauma disrupts normal development and triggers systemic physiological responses, including hormonal surges and immune activation. Recognizing trauma as a multifaceted phenomenon that affects both mind and body is essential to understanding how it becomes biologically embedded.

3. Psychological Effects as a Basis for Biological Change

The psychological consequences of childhood trauma-such as anxiety, depression, dissociation, and impaired emotional regulation-are closely linked to biological alterations. Chronic activation of the brain’s stress response systems leads to sustained cortisol release and other hormonal changes that affect cellular function. This persistent psychological distress acts as a continuous biological stressor, reshaping cellular environments and gene expression patterns throughout the body. Thus, mental suffering and cellular changes are deeply intertwined.

4. The Mind-Body Connection

The intimate relationship between psychological experiences and bodily states means that trauma is not confined to the brain or mind. Activation of the autonomic nervous system, endocrine pathways, and immune responses in response to trauma affects cellular function across multiple organ systems. For example, chronic sympathetic nervous system activation influences heart rate, digestion, and immune surveillance, altering receptor sensitivity and intracellular signaling within cells. This systemic response embeds trauma within the body’s cellular matrix, explaining why trauma survivors often experience physical symptoms such as chronic pain and fatigue.

5. Historical Perspectives on Trauma Storage

Historically, trauma was primarily viewed through psychological frameworks, focusing on repressed memories and unconscious conflicts. However, mid-20th century pioneers like Wilhelm Reich and later Peter Levine emphasized the body’s role in holding trauma, proposing that trauma is encoded in muscular tension and autonomic nervous system patterns. These somatic theories paved the way for modern research into the molecular and cellular mechanisms of trauma storage, bridging psychology and biology and challenging the mind-body dualism.

6. Cellular Memory: Concept and Evidence

Cellular memory refers to the idea that cells retain information about past experiences through molecular changes that persist beyond the initial trauma. This concept is supported by evidence from epigenetics, neurobiology, and immunology. Cells respond to environmental stressors by modifying gene expression, protein synthesis, and metabolism. In trauma, these adaptations can become stable, effectively encoding the history of stress within cells. For instance, immune cells exposed to chronic stress show altered cytokine profiles long after the trauma, while neurons in affected brain regions exhibit changes in synaptic plasticity and gene expression.

7. Epigenetics: The Molecular Language of Trauma

Epigenetics provides a molecular mechanism for how trauma is stored in cells. It involves chemical modifications to DNA and histones that regulate gene activity without changing the DNA sequence. Trauma induces epigenetic changes such as DNA methylation and histone modification, which influence genes involved in stress response, immune function, and neural plasticity. These changes can be stable and long-lasting, persisting throughout life and even across generations, effectively “writing” trauma into the cell’s molecular code.

8. DNA Methylation and Trauma

DNA methylation, the addition of methyl groups to DNA, generally suppresses gene expression. Childhood trauma has been linked to increased methylation of the glucocorticoid receptor gene (NR3C1), which impairs the body’s ability to regulate cortisol, the primary stress hormone. This epigenetic change disrupts the negative feedback loop controlling stress responses, leading to heightened vulnerability to anxiety, depression, and other disorders. Studies of individuals with PTSD and histories of early trauma consistently show distinct methylation patterns in stress-related genes, highlighting DNA methylation’s role in trauma’s cellular imprint.

9. Histone Modification: Remodeling Gene Accessibility

Histones are proteins around which DNA is wrapped, and their chemical modifications alter chromatin structure, influencing gene accessibility. Trauma can induce changes in histone acetylation and methylation that either open or close chromatin regions, regulating expression of genes critical for memory, emotion regulation, and stress resilience. For example, decreased histone acetylation in the hippocampus reduces expression of genes involved in synaptic plasticity, contributing to memory impairments and emotional dysregulation seen in trauma survivors.

10. Telomere Shortening: Trauma and Accelerated Cellular Aging

Telomeres are protective caps at chromosome ends that shorten with each cell division and under stress. Childhood trauma accelerates telomere shortening, effectively hastening biological aging. This premature aging reduces cellular regenerative capacity and increases susceptibility to chronic diseases such as cardiovascular disorders, diabetes, and neurodegenerative conditions. The link between trauma and telomere dynamics illustrates how early adversity translates into long-term health risks through cellular aging mechanisms.

Testimony:
“I struggled with unexplained chronic fatigue and persistent health issues for years. When my doctor explained that my childhood trauma might have ‘aged’ my cells prematurely, it was a revelation. Suddenly, my pain and exhaustion made sense-they were part of my body’s memory.” - Sarah, 42.

11. Neurobiological Consequences: Brain Regions Altered by Trauma

Childhood trauma causes lasting changes in critical brain regions, including the amygdala, hippocampus, and prefrontal cortex. The amygdala, responsible for processing fear and threat, becomes hyperactive, leading to heightened anxiety and hypervigilance. The hippocampus, essential for memory formation and contextualizing experiences, often shows reduced volume, impairing the ability to distinguish past trauma from present safety. The prefrontal cortex, which governs executive functions and emotional regulation, frequently exhibits decreased activity, contributing to difficulties in impulse control and decision-making. These structural and functional changes are reflected at the cellular level through altered synaptic connections, neuroinflammation, and disrupted neurotransmitter systems, all of which perpetuate trauma’s effects.

Testimony:
“Even years after leaving my abusive home, my brain felt like it was stuck on high alert. I couldn’t relax, and memories would flood back unexpectedly. Understanding that my brain’s wiring had changed because of trauma helped me be kinder to myself during recovery.” - James, 35.

12. The Hypothalamic-Pituitary-Adrenal (HPA) Axis: Central Stress Regulator

The HPA axis orchestrates the body’s hormonal response to stress by regulating cortisol release. Childhood trauma disrupts this system, often causing either excessive or blunted cortisol secretion. This dysregulation impairs the body’s ability to adapt to new stressors and affects multiple cellular processes, including metabolism, immune function, and brain plasticity. Persistent HPA axis dysfunction contributes to chronic inflammation, mood disorders, and increased susceptibility to illness, illustrating how trauma’s biological imprint extends into fundamental regulatory systems.

Testimony:
“After years of feeling exhausted and anxious, tests showed my cortisol levels were all over the place. My therapist explained that my childhood trauma had ‘rewired’ my stress system. It was a relief to know it wasn’t just in my head.” - Maria, 29.

13. Cortisol’s Dual Role: Protective and Damaging

Cortisol is vital for managing acute stress, helping the body respond and recover. However, chronic elevation of cortisol, often seen in trauma survivors, damages cells by promoting oxidative stress, impairing mitochondrial function, and triggering inflammation. These cellular damages contribute to the physical health problems frequently observed in those with early trauma histories, such as cardiovascular disease, autoimmune disorders, and metabolic syndrome. The paradox of cortisol’s protective yet potentially harmful role underscores the complexity of trauma’s biological effects.

14. Autonomic Nervous System Dysregulation: Persistent Fight or Flight

Trauma leads to chronic imbalance in the autonomic nervous system, with persistent sympathetic (“fight or flight”) activation and reduced parasympathetic (“rest and digest”) activity. This imbalance affects heart rate variability, digestion, immune surveillance, and cellular signaling pathways. At the cellular level, this translates into altered receptor sensitivities and disrupted intracellular communication, embedding trauma into the physiological rhythms that govern health and behavior.

15. Immune System Alterations: Chronic

Inflammation as Trauma’s Signature
Early life trauma is associated with chronic low-grade inflammation, characterized by elevated pro-inflammatory cytokines such as interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-α). This inflammatory state damages tissues, disrupts metabolic processes, and increases the risk of autoimmune diseases and depression. Immune cells themselves show altered gene expression and function, reflecting a cellular memory of trauma that perpetuates systemic health consequences.

16. Cytokines and Cellular Communication Disruption

Pro-inflammatory cytokines act as messengers between immune cells but, when chronically elevated due to trauma, they disrupt normal cellular communication and repair mechanisms. This dysregulation fosters neuroinflammation and oxidative stress, which impair neuronal function and contribute to mood disorders and cognitive decline in trauma survivors.

17. Mitochondrial Dysfunction: Energy Deficits in Trauma-Exposed Cells

Mitochondria, the cell’s powerhouses, are highly sensitive to stress hormones and inflammatory signals. Childhood trauma impairs mitochondrial function, reducing ATP production and increasing reactive oxygen species. This energy deficit compromises cellular repair and resilience, contributing to symptoms like fatigue, brain fog, and increased vulnerability to chronic illnesses.

18. Metabolic Shifts: Oxidative Stress and Cellular Damage

Trauma-induced stress shifts cellular metabolism toward pathways that generate more reactive oxygen species, leading to oxidative damage of DNA, proteins, and lipids. This damage undermines cellular integrity and accelerates aging processes, further embedding trauma’s effects at the molecular level.

19. Intergenerational Transmission of Trauma: Epigenetic Inheritance

Trauma’s biological imprint can be passed down to subsequent generations through epigenetic modifications in germ cells. Children of trauma survivors often exhibit altered stress responses and increased risk for psychiatric and physical illnesses, even without direct exposure to trauma themselves. This phenomenon highlights trauma’s far-reaching impact beyond the individual.

Testimony:
“My parents never talked about their past, but I struggled with anxiety and depression from a young age. Learning about epigenetic trauma inheritance helped me understand that their pain was, in part, written in my cells.” - Daniel 27

20. Animal Models: Mechanistic Insights into Trauma Inheritance

Animal studlies have provided compelling evidence that trauma-induced epigenetic changes can be passed from parents to offspring. For example, in rodent models, exposure of male mice to chronic stress before mating results in offspring that exhibit altered stress responses, anxiety-like behaviors, and changes in gene expression related to the HPA axis. These changes are mediated by epigenetic marks on sperm DNA, such as DNA methylation and small non-coding RNAs, which influence embryonic development. Such studies highlight the biological plausibility of trauma inheritance and demonstrate that the cellular memory of trauma can transcend generations, affecting descendants who never directly experienced the original stress.

21. Prenatal Stress and Fetal Programming

Prenatal exposure to maternal stress and trauma profoundly impacts fetal development through hormonal and epigenetic mechanisms. Elevated maternal cortisol crosses the placenta, exposing the fetus to high stress hormone levels that can alter brain development, immune system maturation, and metabolic programming. This fetal programming increases the risk for neurodevelopmental disorders, anxiety, depression, and chronic diseases later in life. Epigenetic modifications in fetal tissues, including DNA methylation changes in stress-related genes, provide a molecular mechanism for how prenatal trauma becomes biologically embedded, shaping lifelong health trajectories.

22. The Gut Microbiome: Trauma’s Hidden Mediator

The gut microbiome, the complex community of microorganisms residing in the digestive tract, plays a crucial role in immune regulation, metabolism, and brain function through the gut-brain axis. Early-life trauma has been shown to disrupt the composition and diversity of gut microbiota, leading to dysbiosis. This imbalance contributes to systemic inflammation, altered neurotransmitter production, and impaired stress resilience. Changes in microbial metabolites can influence epigenetic regulation in host cells, further embedding trauma’s effects at the cellular level. Thus, the microbiome acts as a dynamic mediator linking early trauma to systemic biological changes.

23. Connective Tissue and Fascia: Physical Storage of Trauma

Beyond molecular changes, trauma is also stored in the body’s connective tissues, including fascia, muscles, and tendons. Chronic muscular tension and fascial restrictions develop as protective responses to trauma, encoding stress patterns that affect posture, movement, and pain perception. These somatic imprints can persist long after the traumatic event, contributing to chronic pain syndromes and physical dysfunction. The fascia’s dense network of sensory nerve endings and its role in proprioception make it a key site for trauma’s bodily memory, influencing both physical and emotional well-being.

24. Somatic Memory: The Body’s Archive of Trauma

Somatic memory refers to the phenomenon where the body retains trauma-related sensations, emotions, and autonomic patterns independent of conscious recall. Survivors often experience unexplained physical symptoms-such as pain, tension, or numbness-that correspond to unresolved trauma stored in bodily tissues and cells. These somatic memories can be triggered by sensory cues, leading to flashbacks or dissociative states. Understanding somatic memory is essential for trauma therapy, as it highlights the need to address the body alongside the mind in healing.

25. Psychoneuroimmunology: Integrating Mind, Brain, and Body

Psychoneuroimmunology (PNI) is an interdisciplinary field studying how psychological factors influence immune function and overall health. Trauma activates neural and hormonal pathways that modulate immune responses, leading to chronic inflammation and altered cellular function. PNI research demonstrates that stress-induced immune changes are not merely symptoms but active contributors to disease development. This integrative framework explains how trauma’s psychological and biological effects converge at the cellular level, impacting health outcomes.

26. Trauma and Chronic Disease Risk

The biological embedding of childhood trauma increases vulnerability to a wide range of chronic diseases. Persistent inflammation, immune dysregulation, metabolic disturbances, and accelerated cellular aging contribute to heightened risks of cardiovascular disease, type 2 diabetes, autoimmune disorders, and neurodegenerative diseases. Epidemiological studies consistently show that individuals with adverse childhood experiences have higher rates of these conditions, underscoring trauma’s profound impact on long-term health via cellular mechanisms.

27. PTSD and Cellular Dysregulation

Post-Traumatic Stress Disorder (PTSD) exemplifies how trauma disrupts cellular homeostasis. PTSD patients exhibit altered gene expression in immune cells, increased inflammatory markers, and dysregulated cortisol rhythms. Neuroimaging reveals neuroinflammation and reduced hippocampal volume, reflecting cellular and structural brain changes. These biological alterations contribute to the persistence of PTSD symptoms and increased risk for comorbid physical illnesses, highlighting trauma’s deep cellular imprint.

28. Autoimmune Diseases and Trauma-Induced Inflammation

Chronic inflammation resulting from trauma can trigger autoimmune processes, where the immune system mistakenly attacks healthy tissues. Studies link childhood trauma to increased incidence of autoimmune diseases such as rheumatoid arthritis, lupus, and multiple sclerosis. Trauma-induced epigenetic changes in immune cells may alter tolerance mechanisms, promoting autoimmunity. This connection illustrates how trauma’s cellular effects extend beyond mental health to fundamental immune dysregulation.

29. Cardiovascular Disease: Cellular Damage from Trauma

Trauma-related stress elevates cortisol and inflammatory cytokines that damage endothelial cells lining blood vessels. This damage promotes atherosclerosis, hypertension, and increased cardiovascular risk. Cellular oxidative stress and impaired repair mechanisms further exacerbate vascular injury. These biological pathways explain the strong association between early trauma and adult cardiovascular disease observed in epidemiological studies.

30. Neurodegenerative Diseases: Trauma’s Role in Brain Aging

Chronic inflammation and oxidative stress induced by trauma accelerate brain aging processes, increasing vulnerability to neurodegenerative diseases like Alzheimer’s and Parkinson’s. Cellular damage to neurons and glial cells impairs synaptic function and neural plasticity, contributing to cognitive decline. Epigenetic alterations in brain cells may also disrupt gene expression patterns critical for neuronal survival, linking trauma to neurodegeneration at the cellular level.

31. Psychological Symptoms Rooted in Cellular Changes

Anxiety, depression, and emotional dysregulation observed in trauma survivors correlate with cellular and molecular alterations. Neuroinflammation, impaired neurotransmitter systems, and epigenetic modifications in brain cells affect mood regulation circuits. These biological changes provide a mechanistic basis for the psychological symptoms and highlight the importance of addressing cellular health in mental health treatment.

32. Addiction Vulnerability: Trauma Alters Reward Circuits

Childhood trauma reshapes neural reward pathways, altering dopamine signaling and gene expression in the mesolimbic system. These changes increase vulnerability to substance use disorders as individuals seek to self-medicate emotional pain. Cellular adaptations in reward circuits perpetuate addictive behaviors and complicate recovery, emphasizing trauma’s pervasive biological impact.

33. Epigenetic Therapy: Potential to Reverse Trauma’s Effects

Emerging pharmacological interventions targeting epigenetic enzymes, such as histone deacetylase (HDAC) inhibitors, show promise in reversing trauma-induced gene silencing. These therapies aim to restore normal gene expression patterns in brain and immune cells, potentially alleviating symptoms and improving resilience. While still experimental, epigenetic therapy represents a frontier in trauma treatment addressing its cellular roots.

34. Somatic Experiencing: Releasing Stored Trauma

Somatic Experiencing is a body-centered therapeutic approach that helps individuals become aware of and release trauma stored in bodily sensations and autonomic patterns. By gently tracking physical sensations and facilitating completion of defensive responses, this therapy restores autonomic balance and reduces chronic tension. It exemplifies how addressing the body’s cellular memory is essential for healing trauma.

35. Eye Movement Desensitization and Reprocessing (EMDR)

EMDR facilitates the processing and integration of traumatic memories by combining bilateral sensory stimulation with focused recall. Neurobiological studies show that EMDR modulates amygdala activity and strengthens prefrontal regulation, promoting neural plasticity. These effects extend to cellular signaling pathways involved in memory reconsolidation and stress response, illustrating trauma’s biological malleability.

36. Yoga: Enhancing Parasympathetic Tone and Cellular Health

Yoga practice activates the parasympathetic nervous system, reducing inflammation and oxidative stress at the cellular level. Regular yoga enhances heart rate variability, lowers cortisol, and improves mitochondrial function, supporting cellular repair and resilience. Its integrative approach addresses both mind and body, making it effective for trauma recovery.

37. Mindfulness Meditation: Modulating Gene Expression

Mindfulness meditation reduces stress and inflammation by altering gene expression related to immune function and oxidative stress. Studies report decreased pro-inflammatory cytokines and increased expression of genes involved in cellular repair following meditation practice. These molecular changes underpin mindfulness’s therapeutic benefits for trauma survivors.

38. Nutrition: Supporting Cellular Repair

Anti-inflammatory diets rich in antioxidants, omega-3 fatty acids, and polyphenols support cellular health and epigenetic regulation. Proper nutrition enhances mitochondrial function, reduces oxidative damage, and promotes neurogenesis, aiding recovery from trauma’s biological effects.

39. Exercise: Epigenetic Benefits and Mitochondrial Support

Physical activity induces beneficial epigenetic changes that regulate stress response genes and improve mitochondrial biogenesis. Exercise reduces inflammation, enhances neuroplasticity, and improves mood, making it a powerful adjunct in trauma healing.

40. Pharmacological Interventions: Targeting Neurotransmitters and Inflammation

Pharmacological treatments for trauma-related disorders often focus on correcting neurotransmitter imbalances and reducing inflammation. Selective serotonin reuptake inhibitors (SSRIs) and other antidepressants modulate serotonin and norepinephrine pathways, which are frequently dysregulated after trauma. Additionally, anti-inflammatory drugs are being investigated for their potential to alleviate neuroinflammation linked to trauma. Emerging research explores epigenetic drugs that can modify gene expression patterns altered by trauma, offering hope for more targeted and effective therapies that address trauma’s cellular roots rather than just symptoms.

41. Advances in Trauma Research Technologies

Recent technological advances have revolutionized trauma research. Genomic sequencing, epigenome mapping, and single-cell RNA sequencing allow scientists to observe trauma-induced changes at unprecedented resolution. Functional MRI and PET scans reveal how trauma alters brain connectivity and inflammation. Artificial intelligence and machine learning analyze complex datasets to identify biomarkers of trauma exposure and resilience. These tools enable personalized medicine approaches, tailoring interventions based on an individual’s unique biological trauma signature.

42. Ethical Considerations in Trauma Epigenetics

While trauma epigenetics offers exciting possibilities, it raises ethical issues. The potential to identify trauma-related epigenetic marks could lead to stigmatization or discrimination in employment or insurance. Privacy concerns emerge around genetic and epigenetic data. Furthermore, the intergenerational transmission of trauma raises questions about responsibility and consent. Ethical frameworks must balance scientific progress with respect for individual rights and social justice.

43. Trauma-Informed Care: Integrating Biology and Psychology

Trauma-informed care recognizes the pervasive impact of trauma on physical and mental health and integrates this understanding into healthcare practice. Providers trained in trauma-informed approaches create safe environments, avoid retraumatization, and address biological as well as psychological needs. Incorporating knowledge of trauma’s cellular imprint leads to more compassionate, effective care and improved patient outcomes.

44. Social Determinants of Health and Trauma

Social factors such as poverty, discrimination, and community violence exacerbate trauma’s biological effects. Chronic stress from social adversity compounds cellular inflammation, epigenetic dysregulation, and autonomic imbalance. Addressing social determinants is crucial for mitigating trauma’s impact and promoting health equity.

45. Personal Testimonies: The Body Remembers

Many trauma survivors report that their bodies “remember” trauma even when conscious recall is limited. Physical symptoms like chronic pain, muscle tightness, and gastrointestinal distress often persist long after psychological healing begins. These testimonies underscore the importance of addressing trauma at the cellular and somatic levels.

46. Case Study 1: Healing Childhood Abuse with Somatic Therapy

A 38-year-old woman with a history of childhood abuse experienced chronic pain and anxiety resistant to traditional therapy. After engaging in somatic experiencing, she reported gradual release of bodily tension and emotional relief. Functional MRI showed decreased amygdala hyperactivity, and inflammatory markers in blood tests declined. This case illustrates how addressing cellular and somatic trauma can complement psychological healing.

47. Case Study 2: Breaking Intergenerational Trauma Cycles

A family with multiple generations affected by trauma-related disorders participated in a combined epigenetic counseling and trauma-informed psychotherapy program. Over two years, younger members showed improved stress regulation and reduced epigenetic markers of trauma in saliva samples. This example highlights the potential to interrupt trauma’s biological inheritance through targeted interventions.

48. The Role of Social Support in Cellular Healing

Strong social connections buffer the biological effects of trauma by reducing stress hormones and inflammation. Supportive relationships promote parasympathetic activation, enhance neuroplasticity, and improve immune function. Social support is a critical factor in cellular resilience and recovery from trauma.

49. Cellular Markers of Resilience

Not all individuals exposed to trauma develop lasting biological damage. Some show adaptive epigenetic profiles, efficient stress hormone regulation, and robust immune responses. Identifying cellular markers of resilience helps understand protective mechanisms and informs prevention and treatment strategies.

50. The Placebo Effect and Cellular Repair

The placebo effect demonstrates the power of belief and expectation in triggering physiological healing. Placebo responses can modulate gene expression, reduce inflammation, and activate brain regions involved in pain and emotion regulation. This phenomenon highlights the mind’s influence on cellular processes relevant to trauma recovery.

Conclusion

The convergence of psychological insights, cellular biology, and intergenerational studies paints a comprehensive ‹picture of trauma's enduring impact. Recognizing childhood trauma not just as a mental health challenge but as a systemic biological event necessitates a paradigm shift. By understanding how trauma imprints itsellf on our cells, we unlock new avenues for targeted interventions, personalized care, and the potenltial to break cycles of suffering. Embracing this knowledge offers hope for more effective, holistic approaches to healing and a future where early adversity doesn't dictate lifelong outcomes.

References:

1.Van der Kolk, B. A. (2014). The Body Keeps the Score: Brain, Mind, and Body in the Healing of Trauma. Viking.

2.Meaney, M. J., & Szyf, M. (2005). Environmental programming of stress responses through DNA methylation: Life at the interface between a dynamic environment and a fixed genome. Dialogues in Clinical Neuroscience, 7(2), 103–123.

3.Yehuda, R., Bierer, L. M. (2008). Transgenerational transmission of cortisol and PTSD risk. Progress in Brain Research, 167, 121–135.

4.Anda, R. F., Felitti, V. J., et al. (1998). Relationship of childhood abuse and household dysfunction to many of the leading causes of death in adults. American Journal of Preventive Medicine, 14(4), 245-258.

5.Levine, P. A. (1997). Waking the Tiger: Healing Trauma. North Atlantic Books.

A simple and natural wellness secret awaits you. Click here to learn and more change your routine.

https://medicinalseedkit.com/kit/#aff=selja48

0
Subscribe to my newsletter

Read articles from Selja directly inside your inbox. Subscribe to the newsletter, and don't miss out.

Written by

Selja
Selja

Committed to helping you achieve optimal health through practical and natural solutions.