MIT ANALYSIS OF ALTERNATING MOOD DISORDER AND ITS THERAPEUTICS

Alternating Mood Disorder (AMD) encompasses a spectrum of mood disorders characterized by significant fluctuations in an individual’s emotional state, oscillating between periods of manic or hypomanic episodes and depressive episodes. Unlike the more linear trajectory of unipolar depression or the elevated states of standalone mania, AMD involves a cyclic pattern, leading to considerable disruption in the life of the affected individual. This article delves into the etiology, symptoms, diagnosis, and treatment options for AMD, providing a comprehensive overview for both medical professionals and the general public.

The precise causes of AMD remain complex and multifaceted, involving a combination of genetic, neurobiological, and environmental factors. Research suggests a strong genetic component, with individuals having a family history of mood disorders being at higher risk. Neurobiological factors include imbalances in neurotransmitters, such as serotonin, dopamine, and norepinephrine, which play pivotal roles in mood regulation. Environmental stressors, traumatic events, and substance abuse can also trigger or exacerbate symptoms.

The hallmark of AMD is the significant mood oscillation between manic/hypomanic episodes and depressive episodes. Manic/Hypomanic Episodes are characterized by a persistently elevated, expansive, or irritable mood, lasting at least one week for mania or four days for hypomania. Symptoms may include inflated self-esteem, decreased need for sleep, talkativeness, racing thoughts, distractibility, increased goal-directed activity, and excessive involvement in risky behaviours. Depressive Episodes involve pervasive feelings of sadness, hopelessness, or emptiness, with a marked loss of interest or pleasure in most activities. Additional symptoms may include significant weight loss or gain, insomnia or hypersomnia, fatigue, feelings of worthlessness, diminished ability to think or concentrate, and recurrent thoughts of death or suicide.

Diagnosis of AMD requires a careful clinical assessment, including a detailed psychiatric history and a mental status examination. Diagnostic criteria as outlined by the DSM-5 (Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition) or ICD-11 (International Classification of Diseases, 11th Revision) are used to differentiate AMD from other mood disorders. It’s crucial to distinguish between bipolar I disorder, where manic episodes are prominent, and bipolar II disorder, characterized by hypomanic and depressive episodes, as treatment approaches may differ.

Treatment of AMD is multifaceted, aiming to stabilize mood fluctuations, reduce symptom severity, and prevent recurrence. Mood stabilizers (e.g., lithium, valproate) are the cornerstone of treatment, often in conjunction with antipsychotic medications or antidepressants, depending on the nature of the episodes. Cognitive-behavioural therapy (CBT) and interpersonal therapy (IPT) can be effective in addressing thought patterns and social dynamics contributing to mood swings. Regular exercise, adequate sleep, stress management, and avoiding substances that can trigger mood episodes are crucial components of a comprehensive treatment plan. Educating patients and their families about the nature of the disorder, its treatment, and coping strategies is essential for long-term management.

Alternating Mood Disorder presents significant challenges due to its cyclical nature and the impact on various aspects of an individual’s life. However, with accurate diagnosis and a tailored treatment plan, many individuals can achieve substantial improvement and lead fulfilling lives. Ongoing research into the biological and psychological underpinnings of AMD holds promise for even more effective interventions in the future.

PATHOPHYSIOLOGY OF ALTERNATING MOOD DISORDERS

The pathophysiology of Alternating Mood Disorder, particularly bipolar disorder which encompasses bipolar I and II disorders, involves a complex interplay of genetic, neurobiological, and environmental factors. Understanding these underlying mechanisms is crucial for developing effective treatment strategies. Here is a breakdown of the key components involved in the pathophysiology:

There is strong evidence to suggest a genetic component to bipolar disorder. Studies involving twins and families have shown a higher concordance rate among monozygotic twins compared to dizygotic twins, indicating a genetic vulnerability. Specific genetic loci and mutations have been associated with an increased risk, although no single gene is responsible.

Dysregulation of key neurotransmitters, including serotonin, norepinephrine, and dopamine, is central to the mood swings seen in bipolar disorder. For instance, manic episodes are often associated with an excess of norepinephrine and dopamine, while depressive episodes correlate with deficiencies in these neurotransmitters.

Brain imaging studies have identified structural and functional abnormalities in several brain regions in individuals with bipolar disorder. These include the prefrontal cortex, amygdala, hippocampus, and other parts of the limbic system, which are involved in emotion regulation, decision-making, and stress response.

Disruptions in circadian rhythms and sleep-wake cycles are common in bipolar disorder and may contribute to mood swings. The suprachiasmatic nucleus (SCN) of the hypothalamus, which regulates circadian rhythms, may function abnormally in individuals with bipolar disorder, affecting melatonin production, sleep patterns, and mood.

Stressful life events and trauma can trigger episodes of mania or depression in susceptible individuals. The interaction between environmental stressors and genetic predisposition is a key aspect of the disorder’s pathophysiology, with stress potentially altering brain chemistry and functioning.

Abnormalities in the Hypothalamic-Pituitary-Adrenal (HPA) Axis, responsible for the stress response, have been observed in bipolar disorder. Elevated cortisol levels and altered feedback mechanisms can affect mood and behavior, contributing to the cyclical nature of the disorder.

Emerging research suggests a role for inflammation in bipolar disorder. Elevated levels of pro-inflammatory cytokines have been reported during manic and depressive episodes, indicating that immune system dysregulation may play a role in the pathophysiology.

Alterations in ion channels, particularly calcium channels, have been implicated in bipolar disorder. These changes can affect neuronal excitability and neurotransmitter release, leading to mood disturbances. Additionally, abnormalities in intracellular signalling pathways, including those regulated by cyclic adenosine monophosphate (cAMP), have been associated with bipolar disorder.

The pathophysiology of Alternating Mood Disorder is multifaceted and involves a range of biological and environmental components. Understanding these mechanisms is essential for identifying biomarkers for diagnosis and prognosis, as well as developing targeted therapies to manage and treat the disorder. Ongoing research into the genetic, neurobiological, and psychosocial aspects of bipolar disorder continues to shed light on its complex nature.

ENZYME KINETICS IN ALTERNATING MOOD DISORDER

In the context of Alternating Mood Disorders, particularly bipolar disorder, various enzymes play significant roles in neurotransmitter metabolism, signal transduction, and other cellular processes that affect mood regulation. Understanding the enzymes involved, along with their activators and inhibitors, is crucial for developing targeted therapeutic strategies. Here is an overview:

Monoamine Oxidase (MAO) is involved in the catabolism of monoamine neurotransmitters such as dopamine, norepinephrine, and serotonin, which are crucial in mood regulation. Factors that increase oxidative stress can enhance MAO activity, leading to decreased levels of monoamines and potentially contributing to depressive symptoms. MAO inhibitors (MAOIs) such as tranylcypromine and phenelzine act by blocking the activity of MAO, thereby increasing the levels of monoamine neurotransmitters and alleviating symptoms of depression.

Catechol-O-Methyltransferase (COMT) metabolizes catecholamines like dopamine and norepinephrine. It plays a key role in the prefrontal cortex, affecting cognitive functions and mood regulation. Factors that increase the availability of S-adenosylmethionine (SAM), a methyl donor for COMT, can enhance its activity. COMT inhibitors, such as tolcapone and entacapone (more commonly used in Parkinson’s disease for their effect on dopamine metabolism), might influence mood by altering catecholamine levels.

Protein Kinase C (PKC) is involved in signal transduction pathways that regulate a variety of neuronal functions, including neurotransmitter release and receptor sensitivity. Diacylglycerol (DAG) and increased intracellular calcium levels can activate PKC. PKC inhibitors like tamoxifen and lithium (the latter is commonly used in bipolar disorder management) have been shown to have mood-stabilizing effects.

Glycogen Synthase Kinase-3 (GSK-3) is involved in various cellular processes, including modulation of circadian rhythms and neuronal plasticity. It’s implicated in the pathophysiology of bipolar disorder. Pathways involving growth factors and neurotransmitters can activate GSK-3. Lithium is also a well-known inhibitor of GSK-3, contributing to its mood-stabilizing properties by affecting neuroplasticity and possibly reducing neuroinflammation.

Phospholipase C (PLC) plays a role in the phosphoinositide pathway, which is involved in signal transduction in neurons, affecting mood regulation. G protein-coupled receptors (GPCRs) can activate PLC, leading to the production of DAG and inositol triphosphate (IP3), which further participate in cellular signalling pathways. Specific inhibitors of PLC are under research for various indications, and their potential impact on mood disorders is an area of ongoing study.

Adenylyl Cyclase converts ATP to cyclic AMP (cAMP), a second messenger that plays a critical role in the cellular response to hormones and neurotransmitters. GPCRs, upon activation by neurotransmitters, can stimulate adenylyl cyclase activity. Certain mood stabilizers and antipsychotic drugs can indirectly affect adenylyl cyclase activity by modulating receptor function or through downstream effects on signal transduction pathways.

The regulation of these enzymes and their pathways offers potential targets for the treatment of mood disorders. The development of drugs that can more precisely modulate these enzymatic activities holds promise for more effective and tailored therapeutic options for individuals with Alternating Mood Disorders.

ROLE OF DRUGS IN ALTERNATING MOOD DISORDER

Certain medications can trigger or exacerbate symptoms of alternating mood disorders, such as bipolar disorder, by affecting neurotransmitter systems, neuroendocrine pathways, and neural plasticity. Understanding the mechanisms by which these drugs influence mood disorders is crucial for managing patients with a history of or predisposition to such conditions. Here’s a rundown of some notable medications, their mechanisms of action, and how they might influence mood disorders:

Corticosteroids affect the hypothalamic-pituitary-adrenal (HPA) axis and increase the availability of neurotransmitters such as norepinephrine and dopamine, which can lead to mood elevation. They can induce manic-like symptoms, especially with high doses or prolonged use, and may precipitate manic or depressive episodes in susceptible individuals.

Most antidepressants increase the availability of serotonin and/or norepinephrine in the brain. Selective serotonin reuptake inhibitors (SSRIs), for example, specifically block the reuptake of serotonin. While effective for depressive episodes, antidepressants can trigger manic or hypomanic episodes in individuals with bipolar disorder, especially if used without a mood stabilizer.

Stimulants such as amphetamines and methylphenidate increase the release of norepinephrine and dopamine, enhancing alertness, attention, and energy. These medications can exacerbate or trigger manic symptoms or contribute to mood instability, particularly in those with an underlying mood disorder.

Atypical antipsychotics block dopamine and serotonin receptors, which can stabilize mood from a high state. However, their effect on the dopaminergic and serotonergic systems can be complex. While often used to treat manic episodes, some antipsychotics can lead to depressive symptoms due to their dampening effect on dopamine pathways.

Interferons, used primarily for treating certain cancers and viral infections, interferons can alter immune function and neurotransmitter levels, contributing to inflammation and affecting mood regulation. Treatment with interferons has been associated with the onset of depressive symptoms and, less commonly, mood elevation or instability.

Overreplacement or aggressive treatment of hypothyroidism with thyroid hormones (e.g., levothyroxine) can elevate thyroid hormone levels, affecting metabolism and neurotransmitter activity. Excessive thyroid hormone supplementation can induce symptoms of hyperthyroidism, including mood swings, irritability, and even manic episodes.

Substances like cocaine, amphetamines, and alcohol alter neurotransmitter levels rapidly and profoundly. Cocaine and amphetamines increase dopamine and norepinephrine, while alcohol primarily affects the GABAergic system but also impacts dopamine and serotonin. These substances can cause significant mood dysregulation, inducing manic or depressive episodes in susceptible individuals.

ROLE OF PHYTOCHEMICALS IN ALTERNATING MOOD DISORDER

The impact of phytochemicals—naturally occurring compounds found in plants—on mood disorders is a complex and emerging field of study. Some phytochemicals may influence mood and cognition, potentially exacerbating symptoms in individuals with alternating mood disorders, such as bipolar disorder. It’s crucial to understand that while the consumption of these compounds in a typical diet is unlikely to cause significant mood alterations, concentrated doses found in supplements or extracts can have more pronounced effects. Here’s a look at several phytochemicals, their mechanisms of action, and how they might influence mood disorders:

Caffeine acts as a central nervous system stimulant by antagonizing adenosine receptors. Adenosine normally promotes sleep and suppresses arousal; by blocking its action, caffeine increases alertness and can elevate mood. In susceptible individuals, excessive caffeine intake can lead to anxiety, sleep disturbances, and mood swings. In those with bipolar disorder, it might contribute to manic episodes or exacerbate anxiety and insomnia during depressive phases.

Tetrahydrocannabinol (THC), the psychoactive component of cannabis, exerts its effects primarily through partial agonism of the cannabinoid receptors CB1 and CB2 in the brain, affecting the release of various neurotransmitters and modulating mood and perception. While some individuals may experience mood stabilization at lower doses, high doses or chronic use can aggravate or trigger symptoms of mania, depression, or mood instability, particularly in those predisposed to mood disorders.

Hyperforin and Hypericin, found in St. John’s Wort, hyperforin is believed to act as a reuptake inhibitor for several neurotransmitters, including serotonin, dopamine, and norepinephrine, similar to antidepressants. Hypericin may contribute to the plant’s overall antidepressant effects. Though used for mild to moderate depression, St. John’s Wort can induce manic episodes in people with bipolar disorder and interact with a wide range of medications, potentially affecting mood stability.

Salvinorin A, the active component of Salvia divinorum, is a potent kappa-opioid receptor agonist. It affects perception, consciousness, and mood by altering neurotransmitter systems in the brain. Its use can lead to significant alterations in mood and perception, including dysphoria and anxiety in some cases, which could exacerbate symptoms in individuals with mood disorders.

Resveratrol, found in grapes and red wine, has antioxidant and anti-inflammatory properties. It may also modulate neurotransmitter systems and neuroendocrine functions, contributing to its potential mood-regulating effects. While often considered beneficial for its antioxidant properties, the impact of resveratrol on mood disorders is not well understood. Theoretical concerns suggest that, in high doses, its estrogenic activity could influence mood swings.

Capsaicin, the spicy component of chili peppers, interacts with the vanilloid receptors, which are involved in pain sensation and possibly mood regulation through endorphin release. While capsaicin might have mood-elevating effects due to pain-induced endorphin release, excessive intake could potentially contribute to anxiety or mood instability in sensitive individuals.

IMMUNOLOGICAL FACTORS IN ALTERENATING MOOD DISORDER

The role of immunological factors in alternating mood disorders, such as bipolar disorder, has gained increasing attention in psychiatric research. This interest stems from the growing understanding that the immune system and the central nervous system (CNS) interact in complex ways that can affect mood regulation. Several immunological factors, including cytokines, autoimmunity, and chronic inflammation, have been implicated in the pathophysiology of mood disorders. Here’s how these factors might play a role:

Cytokines are small signaling proteins released by immune cells that have profound effects on brain function, including neurotransmitter metabolism, neuroendocrine function, and neural plasticity. Pro-inflammatory cytokines, such as interleukin-6 (IL-6), tumor necrosis factor-alpha (TNF-α), and interleukin-1 beta (IL-1β), have been found in elevated levels in some individuals experiencing episodes of mania or depression. These cytokines can cross the blood-brain barrier and interact with the CNS, potentially leading to alterations in mood and behavior. For example, they can affect the metabolism of serotonin and dopamine, neurotransmitters closely associated with mood regulation. Chronic inflammation might also contribute to neuroprogression, the progressive changes in brain structure and function seen in mood disorders.

Some evidence suggests that autoimmune processes, where the body’s immune system mistakenly attacks its own cells, might be linked to the development of certain mood disorders. Autoantibodies targeting CNS structures could alter neural circuits involved in mood regulation. The presence of autoantibodies or an increased prevalence of autoimmune diseases in patients with bipolar disorder suggests an immunological contribution to mood dysregulation. However, the exact mechanisms by which autoimmune processes might contribute to mood disorders are still under investigation.

Microglia are the primary immune cells of the CNS and play a key role in immune surveillance and neuroinflammation. In response to various triggers, microglia can become activated and release cytokines and other inflammatory mediators. Chronic microglial activation has been associated with neuroinflammatory processes that could contribute to the pathophysiology of mood disorders. Activated microglia might not only influence neuroinflammation but also contribute to neuronal damage and synaptic pruning, affecting mood regulation.

The gut-brain axis refers to the bidirectional communication between the gastrointestinal tract and the CNS, involving neural, hormonal, and immunological pathways. Changes in the gut microbiota can influence systemic inflammation and, in turn, brain function and mood. Dysbiosis, or an imbalance in the gut microbiome, has been linked to increased levels of systemic inflammation and might contribute to the onset or exacerbation of mood disorders through the production of inflammatory cytokines.

The understanding that immunological factors can contribute to alternating mood disorders opens new avenues for treatment. Anti-inflammatory drugs, immune modulators, and interventions aimed at reducing systemic inflammation (such as lifestyle modifications to improve diet and gut health) are being explored as potential strategies for managing mood disorders. Moreover, this perspective supports a more holistic approach to treatment, emphasizing the importance of physical health and immune system regulation in maintaining mental health.

HEAVY METALS AND MICROELEMENTS IN ALTERNATING MOOD DISORDER

Heavy metals and certain microelements, when present in excessive or deficient amounts, can have profound effects on mental health, potentially causing or aggravating alternating mood disorders such as bipolar disorder. These elements can interfere with neurobiological pathways, neurotransmitter systems, and oxidative stress mechanisms, among others. Understanding their impact is crucial for both prevention and treatment. Here is an overview of some relevant heavy metals and microelements:

Lead exposure can damage the nervous system by disrupting calcium homeostasis, mimicking calcium, and thus affecting neurotransmitter release and synaptic function. It also induces oxidative stress, damages mitochondrial function, and alters the expression of genes related to synaptic plasticity. Chronic lead exposure has been associated with cognitive deficits, depression, and anxiety. While direct links to bipolar disorder are less clear, the neurotoxic effects of lead could contribute to mood dysregulation.

Mercury can cross the blood-brain barrier and cause neurotoxicity through several mechanisms, including oxidative stress, disruption of calcium homeostasis, and impairment of neurotransmitter systems (e.g., serotonergic, dopaminergic, and cholinergic systems). Exposure to high levels of mercury has been linked to mood swings, irritability, and depression. Its role in exacerbating mood disorders stems from its widespread effects on brain function.

Cadmium exposure leads to oxidative stress, disruption of neurotransmitter systems, and interference with nutrient absorption, such as zinc, a crucial element for brain health. Cadmium has been implicated in an increased risk of depression, and by extension, could influence the course of mood disorders by exacerbating underlying neurobiological disturbances.

Zinc acts as a neurotransmitter modulator, playing roles in synaptic transmission, neurogenesis, and neural plasticity. It also has antioxidant properties and is essential for the function of numerous enzymes. Zinc deficiency has been associated with depressive symptoms and may influence the efficacy of antidepressant therapies. Its role in mood regulation suggests that imbalance could affect the course of mood disorders.

Selenium is crucial for antioxidant defense systems and thyroid hormone metabolism. It influences mood and cognitive function by protecting against oxidative damage and supporting endocrine function. Low selenium levels have been linked to increased risk of depression and other mood disorders, highlighting its importance in mood regulation.

Copper is involved in neurotransmitter synthesis and function, including dopamine and norepinephrine, which are key to mood regulation. However, excess copper can lead to oxidative stress and neurotoxicity. Elevated copper levels have been associated with symptoms of depression and may play a role in mood disorders by disrupting neurotransmitter balance and promoting oxidative stress.

The relationship between heavy metals, microelements, and mood disorders underlines the importance of maintaining a balanced intake and minimizing exposure to toxic metals. This includes dietary management, avoiding known sources of heavy metal exposure, and possibly using supplements under medical supervision for deficiencies.

MIT APPROACH TO ALTERNATING MOOD DISORDER

MIT or Molecular Imprints Therapeutics refers to a scientific hypothesis that proposes a rational model for biological mechanism of homeopathic therapeutics. According to MIT hypothesis, potentization involves a process of ‘molecular imprinting’, where in the conformational details of individual drug molecules are ‘imprinted or engraved as hydrogen- bonded three dimensional nano-cavities into a supra-molecular matrix of water and ethyl alcohol, through a process of molecular level ‘host-guest’ interactions. These ‘molecular imprints’ are the active principles of post-avogadro dilutions used as homeopathic drugs. Due to ‘conformational affinity’, molecular imprints can act as ‘artificial key holes or ligand binds’ for the specific drug molecules used for imprinting, and for all pathogenic molecules having functional groups ‘similar’ to those drug molecules. When used as therapeutic agents, molecular imprints selectively bind to the pathogenic molecules having conformational affinity and deactivate them, thereby relieving the biological molecules from the inhibitions or blocks caused by pathogenic molecules.

According to MIT hypothesis, this is the biological mechanism of high dilution therapeutics involved in homeopathic cure. According to MIT hypothesis, ‘Similia SimilibusCurentur’ means, diseases expressed through a particular group of symptoms could be cured by ‘molecular imprints’ forms of drug substances, which in ‘molecular’ or crude forms could produce ‘similar’ groups of symptoms in healthy individuals. ‘Similarity’ of drug symptoms and diseaes indicates ‘similarity’ of pathological molecular inhibitions caused by drug molecules and pathogenic molecules, which in turn indicates conformational ‘similarity’ of functional groups of drug molecules and pathogenic molecules. Since molecular imprints of ‘similar’ molecules can bind to ‘similar ligand molecules by conformational affinity, they can act as the therapeutics agents when applied as indicated by ‘similarity of symptoms. Nobody in the whole history could so far propose a hypothesis about homeopathy as scientific, rational and perfect as MIT explaining the molecular process involed in potentization, and the biological mechanism involved in ‘similiasimilibus- curentur, in a way fitting well to modern scientific knowledge system.

If symptoms expressed in a particular disease condition as well as symptoms produced in a healthy individual by a particular drug substance were similar, it means the disease-causing molecules and the drug molecules could bind to same biological targets and produce similar molecular errors, which in turn means both of them have similar functional groups or molecular conformations. This phenomenon of competitive relationship between similar chemical molecules in binding to similar biological targets scientifically explains the fundamental homeopathic principle Similia Similibus Curentur.

Practically, MIT or Molecular Imprints Therapeutics is all about identifying the specific target-ligand ‘key-lock’ mechanism involved in the molecular pathology of the particular disease, procuring the samples of concerned ligand molecules or molecules that can mimic as the ligands by conformational similarity, preparing their molecular imprints through a process of homeopathic potentization up to 30c potency, and using that preparation as therapeutic agent.

Since individual molecular imprints contained in drugs potentized above avogadro limit cannot interact each other or interfere in the normal interactions between biological molecules and their natural ligands, and since they can act only as artificial binding sites for specific pathogenic molecules having conformational affinity, there cannot by any adverse effects or reduction in medicinal effects even if we mix two or more potentized drugs together, or prescribe them simultaneously- they will work.

As per the understanding derived from the above discussions, following drugs in homeopathic 30C potentized forms are recommended in the MIT Homeopathy therapeutics of Alternating Mood Disorder. These drugs could be used selecting according to specific indications, or as a combination of multiple drugs. Since molecular imprints cannot interact each other, or produce an harmful effects, it will be more convenient and effective to use in combinations.

Lithium 30, Serotonin 30, Dopamine 30, Adrenalin 30, Interleukin-1 beta (IL-1β)30, Cuprum Met 30, Selenium 30, Zincum Met 30, Cadmium 30, Plumbum Met 30, Mercurius 30, Capsicum 30, Resveratrol 30, Salvia Officinalis 30, Hypericum 30, Cannabis Indica 30, Coffea Crudum 30, Tolcapole 30,




Author: Chandran Nambiar K C

I am Chandran Nambiar K C Author, REDEFINING HOMEOPATHY Managing Director, Fedarin Mialbs Private Limited Developer. SIMILIMUM ULTRA Homeopathic Software I am not a scientist, academician, scholar, professional homeopath or anybody with 'big credentials', but an old lay man, a retired government servant, who accidentally happened to fall into the deep waters of the great ocean of homeopathic knowledge during his fiery teenage years, and was destined to live a whole life exploring the mysteries of that wonderful world with unending enthusiasm. My interest in homeopathy happened very accidentally when I was only 20 years old UNDERGRADUATE ZOOLOGY student, through a constant relationship with a local practitioner who happened to be father of my classmate. I was a regular visitor in his clinic, where from I started reading BOERICKE MATERIA MEDICA and other homeopathic books, which helped me to cure myself my troublesome asthma that have been haunting me since my childhood days. I became a voracious reader of homeopathy.

Leave a comment