Dihydroberberine: A Promising Agent in Metabolic Regulation and Cellular Senescence Reduction

Dihydroberberine (DHB), a chemically modified derivative of Berberine, is gaining recognition for its enhanced bioavailability and significant therapeutic potential, particularly in metabolic regulation and aging. This review highlights DHB's role in targeting metabolic health and reducing cellular senescence through AMPK activation and mTOR pathway modulation. By enhancing AMPK activity, DHB improves insulin sensitivity, lipid metabolism, and inhibits mTORC1, thereby promoting autophagy and diminishing cellular senescence. The article explores the synergistic effects of combining DHB with Rapamycin, an mTOR pathway inhibitor, underscoring their potential to mitigate Rapamycin's metabolic side effects while amplifying its healthspan-extending benefits. Through a detailed examination of molecular mechanisms and comparative studies, this review underscores DHB's promising role in enhancing metabolic health and longevity, presenting it as a potential advancement in therapeutic strategies aimed at combating age-related diseases.

27 mins

By: Shreshtha Jolly

Introduction

Dihydroberberine (DHB) is emerging as a popular supplement for its enhanced bioavailability and therapeutic efficacy compared to its precursor, Berberine. Berberine, a natural compound, has been widely used for its antimicrobial and anti-inflammatory properties. It has also shown promise in improving glucose and lipid metabolism, supporting cardiovascular health, and offering neuroprotective effects.

In this article, we will examine DHB, its potential benefits, and the scientific evidence supporting its use. Healthspan physicians often recommend Berberine in combination with Rapamycin to amplify health benefits, and we will explore the origins and rationale behind this recommendation. By delving into the mechanisms of action, comparative studies, and potential therapeutic applications, we aim to provide a comprehensive overview of DHB and its role in promoting health and longevity.

An Overview of Berberine and Dihydroberberine

Before we delve into the longevity science of Berberine and the differences between Berberine and Dihydroberberine, let's briefly overview the range of use cases of Berberine. While these are interesting use cases, the underlying mechanism of how Berberine works is even more important to understand. Specifically, it targets key Hallmark of Aging pathways that are at the core of these pathologies—loss of proteostasis, deregulated nutrient sensing, mitochondrial dysfunction, cellular senescence, inflammaging, and disabled macroautophagy.

Berberine is a natural compound found in several plant species, including Coptis chinensis (Chinese goldthread), goldenseal, and several Berberis species. Berberine has been widely used as a natural supplement due to its myriad health benefits. It is used to treat various bacterial and fungal infections, including diarrhea, dysentery, and gastrointestinal infections, due to its potent antimicrobial activities [1]. It also provides anti-inflammatory properties, making it useful for treating conditions such as asthma, arthritis, skin infections, and inflammatory bowel disease [2].

Berberine’s anti-inflammatory effects were studied in mouse models of asthma, a chronic lung disease that inflames and narrows the airways, making breathing difficult. Researchers created a mouse model of asthma by exposing mice to ovalbumin, which triggers an inflammatory response in the lungs. Mice treated with the compounds experienced reduced inflammation and swelling in the airways compared to those that did not receive the treatment. This study provides evidence that natural plant-based compounds such as Berberine may have therapeutic potential for inflammatory diseases like asthma [2].

Besides its anti-inflammatory properties, Berberine has also been found to have beneficial effects on glucose and lipid metabolism. It is used to treat conditions such as hyperglycemia (high blood sugar levels that exceed the normal range) and hyperlipidemia (excess fat, particularly cholesterol, in the bloodstream) [3]. 

In the cardiovascular and neurological domains, Berberine continues to show promise. It supports cardiovascular health and helps prevent conditions like hypertension and atherosclerosis—a condition where fatty plaque builds up inside blood vessels, restricting blood flow and increasing susceptibility to heart attacks and strokes. 

Berberine also offers neuroprotective effects, providing defense against neurological disorders such as Alzheimer's and Parkinson's disease [4, 5]. Additionally, some studies suggest that Berberine may have anti-tumor and chemopreventive effects, making it a potential complementary therapy in cancer management.

From the above discussion, it is evident that Berberine offers a range of beneficial effects. However, the compound has limited bioavailability, meaning that our bodies cannot easily absorb and utilize it. To address this issue, researchers have chemically modified the structure of the compound to create Dihydroberberine (DHB). A detailed comparison of the bioavailability and therapeutic efficacy of the two compounds will be presented in the following section.

Comparing Berberine and Dihydroberberine

The conversion of Berberine into Dihydroberberine (DHB) offers several significant benefits that enhance its therapeutic potential. We will delve into this in more detail, specifically focusing on how DHB more effectively activates AMPK, an enzyme central to Berberine’s mechanism of action.

Furthermore, transforming Berberine into DHB also makes the compound safer and more tolerable for individuals. Berberine, in its original form, can sometimes cause gastrointestinal discomfort, including cramping, diarrhea, and constipation. In contrast, DHB is designed to be gentler on the digestive system, reducing these side effects and making it more suitable for long-term use.

Lastly, the most important benefit of this conversion is the significant increase in bioavailability. Bioavailability refers to how well a substance is absorbed and utilized by the body. While Berberine has relatively low bioavailability, meaning that only a small fraction of the ingested compound is absorbed into the bloodstream, DHB has been shown to have much higher bioavailability.

Several studies have underscored the superiority of Dihydroberberine (DHB) over Berberine in managing health outcomes. For instance, a study titled Absorption Kinetics of Berberine and Dihydroberberine and Their Impact on Glycemia: A Randomized, Controlled, Crossover Pilot Trial investigated the bioavailability and therapeutic efficacy of Berberine compared to DHB in a cohort of five healthy adult males. Participants underwent four treatments in random order—placebo, 500 mg of Berberine (B500), 100 mg of DHB (D100), and 200 mg of DHB (D200). They received three of these doses with meals the day before a blood test, and the final dose the next morning with a standardized test meal consisting of a 30 g glucose solution and three slices of white bread. Blood samples were collected at 0, 20, 40, 60, 90, and 120 minutes after ingestion and analyzed for Berberine levels, glucose, and insulin [6].

Results showed that DHB led to significantly higher concentrations in the blood (3.76 ng/mL with D100 and 12.0 ng/mL with D200) compared to Berberine (0.4 ng/mL with B500). This indicates that DHB has better bioavailability and absorption, entering the bloodstream more efficiently and maintaining higher levels for a longer duration. This suggests that DHB may be more effective at lower doses, potentially reducing the risk of gastrointestinal side effects. However, no significant differences in glucose and insulin levels were observed among the groups, likely due to the short duration of the supplementation. Follow-up efficacy studies are needed to assess the impact of these supplements on glucose and insulin levels over a longer period [6].

Similarly, an additional study, titled Comparative pharmacokinetics and safety assessment of transdermal Berberine and dihydroberberine, evaluated the bioavailability and safety profiles of Berberine and DHB in rats. In the study, the researchers used a transdermal formulation of both compounds. They steered away from oral dosages as these have been previously shown to be less bioavailable, requiring higher dosages to achieve desired effects but with added gastrointestinal side effects. 

Overall, they found that the transdermal formulations, especially DHB, resulted in much higher levels of Berberine in the body compared to oral Berberine. Overall, this study supports the safety and improved bioavailability of transdermal DHB compared to oral Berberine. These transdermal compositions have the potential to be more effective in treating conditions like high cholesterol and hyperlipidemia [7].

Why is Dihydroberberine More Bioavailable?

The superior bioavailability of DHB relative to Berberine has to do with the chemical modifications introduced in the former compound. Berberine is hydrogenated to produce DHB. In simple terms, this means that additional hydrogen atoms are introduced to the compound. When this happens, the chemical structure of Berberine is altered in a way that makes it more fat-soluble or lipophilic. This increased lipophilicity allows DHB to more easily cross biological membranes, including our gastrointestinal tract, and enter the bloodstream to mediate its effects.

Overall, the process of converting Berberine into DHB offers several important benefits. Multiple studies have demonstrated the superiority of DHB over Berberine in terms of bioavailability and subsequent health outcomes. Unlike Berberine, DHB's increased lipophilicity improves intestinal absorption and increases plasma concentrations of the active ingredient. This suggests that DHB may be more effective at lower doses, potentially reducing the risk of gastrointestinal side effects associated with higher Berberine dosages.

Berberine and Hallmark of Aging Pathways

The breadth of impact of Berberine is clear when you look at the number of Hallmark of aging pathways it targets. Its effects touch upon multiple hallmarks of aging, including loss of proteostasis, deregulated nutrient sensing, mitochondrial dysfunction, cellular senescence, inflammaging, and impaired autophagy.

Let's examine some foundational studies on the role of Berberine and cellular senescence.

Berberine and Cellular Senescence

As cells age, they often enter a state of senescence, ceasing to divide and secreting inflammatory factors that can damage nearby cells. Recent studies have highlighted the potential of Berberine to target senescence as a driver of aging. A significant contribution to this field was made by Professor Hong Zhao from the Brander Cancer Research Institute at New York Medical College. The group's study, titled Berberine suppresses gero-conversion from cell cycle arrest to senescence, focuses on the effects of Berberine on stress-related cellular senescence, providing valuable insights into its mechanisms and potential applications.

To fully appreciate the findings from Professor Zhao's study, it's crucial to understand cellular senescence, a phenomenon where cells cease to divide and grow as they age. There are two primary forms of cellular senescence: replicative and stress-induced.

Replicative Senescence

Replicative senescence occurs due to the shortening of telomeres, the protective caps at the ends of chromosomes that wear away slightly with each cell division. This form of senescence is triggered when telomeres reach a critically short length, a point known as the Hayflick Limit. At this threshold, cells lose their ability to divide further and enter a state of dormancy. While these cells no longer proliferate, they remain metabolically active and can secrete various substances that influence aging and tissue function.

Stress-induced Senescence (Premature Senescence):

Unlike replicative senescence, stress-induced senescence can occur independently of telomere length and is triggered by external stressors such as oxidative stress, radiation, exposure to chemotherapeutic drugs, or chronic inflammation. These stressors cause damage that the cell cannot repair, leading to a cessation of cell division and changes in cell behavior that can contribute to aging and disease progression.

In the Zhao study, researchers evaluated whether Berberine could offer protective effects in cells exposed to mitoxantrone, a DNA-damaging drug. Mitoxantrone disrupts the DNA replication process as cells divide, predisposing them to a state of premature senescence. 

The research team exposed cells to mitoxantrone to induce premature senescence and then treated these cells with Berberine to assess its potential protective effects. The objective was to determine whether Berberine could mitigate the senescence burden triggered by the DNA damage. The findings were quite revealing; cells treated with Berberine showed a significant reduction in the markers of cellular senescence compared to those only exposed to mitoxantrone. This indicated that Berberine has potent geroprotective properties that can help shield cells from the adverse effects of DNA damage.

To understand how Berberine reduced the overall levels of senescence we have to focus the role of a key regulator of cell growth, mTOR and how its overactivity leads to the acceleration of senescent cell propagation and harm.

Berberine and the mTOR Pathway

The mammalian target of rapamycin (mTOR) is a pivotal enzyme in cell biology, acting much like an air-traffic controller for cellular growth and metabolism. mTOR integrates signals from nutrients, growth factors, and environmental cues to regulate key cellular processes. When conditions are favorable—signaled by an abundance of nutrients and growth factors—mTOR is activated. This activation prompts the cell to produce proteins and undertake cell growth and division, processes vital for tissue repair and growth, particularly in developing organisms.

However, mTOR's role extends beyond just stimulating growth. In conditions where nutrients are sparse, the activity of mTOR naturally declines. This reduction is critical from an evolutionary standpoint, as it adjusts the cell’s priorities under nutrient scarcity. Instead of promoting growth, which is energy-intensive, the cell shifts its focus towards survival. Here, the decrease in mTOR activity triggers the cell's autophagy machinery, a built-in recycling system that breaks down non-essential or damaged cellular components. This process not only conserves resources but also provides the cell with essential nutrients and energy, thus maintaining energy balance and enhancing cellular survival during challenging times.

While mTOR is crucial for cellular growth and response to environmental cues, its chronic overactivation is associated with several age-related diseases. In the aging process, mTOR may not downregulate appropriately, remaining persistently active. This continued activity can lead to uncontrolled cellular growth, a key factor in the development of cancer. At the same time, persistent mTOR activation can hinder the cell's natural repair mechanisms, compounding the damage and dysfunction associated with aging.

Chronic overactivation of mTOR, whether due to excessive nutrient availability, stress, or genetic mutations, can accelerate the aging process. Overactive mTOR signaling leads to rampant cell growth and proliferation, straining and eventually depleting cellular resources. This depletion results in nutrient shortages within the cell, contributing to overall cellular dysfunction and the aging phenotype. The implications are particularly severe in senescent cells—older cells that have stopped dividing but remain metabolically active. Elevated mTOR activity in these cells enhances their inflammatory and growth-stimulating properties.

This pathological behavior in senescent cells can be detrimental, as it may promote the spread of cellular senescence throughout the tissue, exacerbating tissue dysfunction and aging. The "senescence-associated secretory phenotype" (SASP), characterized by the secretion of inflammatory cytokines, chemokines, and growth factors, can disrupt local tissue environments and further drive the aging process and the progression of age-related diseases.

The Role of mTOR and Autophagy

mTOR signaling is a key regulator of autophagy, the cellular process that serves as a critical quality control mechanism. Autophagy ensures cellular health by removing damaged proteins and other cellular debris. It allows cells to recycle and eliminate dysfunctional components efficiently, which is essential for maintaining cellular vitality and function. However, when mTOR signaling becomes dysregulated—often due to genetic factors, environmental stressors, or chronic disease conditions—it can suppress autophagy. This suppression impairs the cell's ability to clear out harmful materials, leading to the accumulation of cellular waste products. Over time, this buildup contributes significantly to cellular dysfunction and accelerates the aging process.

Impaired autophagy is notably significant in the context of age-related diseases, especially in neurodegenerative disorders. In healthy neuronal cells, autophagy plays a vital role in clearing misfolded proteins, such as Tau proteins, which are implicated in neurodegenerative diseases like Alzheimer's. Effective autophagy maintains neuronal health by preventing the accumulation of these toxic proteins. However, in Alzheimer's disease, impaired autophagy results in an excessive buildup of Tau proteins, which disrupts normal cellular functions and triggers an inflammatory immune response that can lead to neuronal cell death [8].

The failure of autophagy to remove these misfolded proteins exemplifies the profound negative consequences that can arise when this essential process is compromised. In the brain, the accumulation of Tau proteins not only promotes inflammation but also activates immune cells, both of which contribute to the neurodegeneration seen in Alzheimer’s disease. This situation highlights the crucial role of autophagy in maintaining neuronal health and preventing the progression of neurodegenerative diseases.

While the consequences of impaired autophagy are starkly evident in neurodegenerative diseases, this fundamental cellular process also plays a critical role in other age-related conditions, including cardiovascular diseases, cancer, and metabolic disorders. In these diseases, just as in neurodegeneration, defective autophagy leads to the buildup of damaged proteins and organelles, exacerbating cellular dysfunction and accelerating disease progression.

In the cardiovascular system, for example, efficient autophagy is crucial for the maintenance of heart muscle cells and blood vessels. Impaired autophagy in these cells can result in the accumulation of dysfunctional mitochondria and other cellular debris, contributing to heart disease and increased susceptibility to myocardial infarction (heart attack).

Similarly, in muscle cells, proper autophagy is essential for removing defective mitochondria and other cellular waste. When autophagy is impaired, these components accumulate, leading to muscle weakness and degeneration—a common feature in sarcopenia and other muscle-wasting conditions associated with aging.

mTOR, Mitochondrial Function and Oxidative Stress

mTOR signaling plays a pivotal role in cellular regulation, but when it becomes dysregulated, one of the significant consequences is an increase in oxidative stress. Activation of mTOR can enhance the production of reactive oxygen species (ROS), which are chemically reactive molecules containing oxygen. Elevated levels of ROS can cause substantial cellular damage, triggering inflammatory responses that further exacerbate cellular aging and contribute to the progression of age-related diseases.

This heightened oxidative stress damages cellular structures and activates various inflammatory pathways. Chronic inflammation, commonly referred to as "inflammaging," is recognized as a hallmark of aging. It is linked to the progression of multiple age-related diseases, including cardiovascular diseases, neurodegenerative disorders, and cancer. The activation of these inflammatory responses by ROS creates a harmful cycle that accelerates the aging process by continuously inflicting damage on the cellular machinery.

Moreover, the influence of mTOR signaling extends to mitochondrial function — the powerhouses of cells that produce energy. Dysregulated mTOR activity can lead to impaired mitochondrial function, characterized by decreased energy production and an increase in ROS generation. Mitochondria, when dysfunctional, may leak electrons during the process of electron transport, which then react with oxygen to form more ROS. This not only exacerbates oxidative stress but also causes damage to mitochondrial DNA, proteins, and lipids.

This cascade of mitochondrial dysfunction and excessive ROS production forms a feedback loop that further accelerates cellular aging. As mitochondria become increasingly damaged, they contribute less effectively to cellular energy production and more to cellular stress and damage. The accumulation of these damaged mitochondria and the resultant oxidative stress can activate cell death pathways or induce cellular senescence—a state where cells cease to divide and begin to secrete inflammatory factors that contribute to tissue dysfunction.

The Effects of Berberine on Senescence in the Zhao Study

Given the significant role that dysregulated mTOR signaling plays in cellular senescence and, consequently, longevity, the Zhao study focused on the connection between Berberine and DHB and the reduction of mTOR activity. 

The study found that the anti-senescence effects of Berberine are closely tied to its ability to inhibit the mTOR pathway. Berberine achieves these effects partly through its interaction with mitochondria, the cell's energy-producing organelles. 

It localizes within mitochondria and influences their function by inhibiting the respiratory electron chain—a series of reactions crucial for energy production. This inhibition leads to a decrease in cellular energy production, which in turn activates AMP-activated protein kinase (AMPK), a cellular energy sensor that helps maintain energy homeostasis.

AMPK activation plays a critical role in how Berberine mediates its anti-aging effects. When activated, AMPK promotes catabolic processes that generate ATP (the energy currency of the cell) and inhibits anabolic processes that consume ATP. Crucially, one of the key actions of activated AMPK is the inhibition of the mTOR signaling pathway. By inhibiting mTOR, AMPK activation leads to a reduction in cellular growth and proliferation signals.

The attenuation of replication stress-induced cellular senescence by Berberine, as observed in the Zhao study, is likely mediated through this AMPK-mTOR pathway. 

In the Zhao study, researchers explored how Berberine decreases cellular senescence by targeting the AMPK-mTOR pathway [9]. Ultimately there were three major contributors to the reduction of senescence:

  • Downregulation of mTOR Activity: Berberine treatment effectively reduced the activity of mTOR. By inhibiting mTOR, Berberine interrupted the signaling cascade that drives excessive cell growth and proliferation, processes often associated with cellular senescence.

  • Reducing of Oxidative Stress: Berberine improves mitochondrial function and efficiency, thereby reducing mitochondrial ROS production. This is particularly important as mitochondria are significant sources of ROS in cells. This reduction in oxidative stress helped to preserve cellular integrity and function, ultimately leading to a decrease in senescence-associated alterations.

  • Increased Autophagy: Berberine was also shown to influence autophagy. The researchers found that Berberine promoted autophagy, leading to enhanced clearance of cellular debris and dysfunctional components. This could then contribute to the suppression of cellular senescence.

The inhibition of mTOR by Berberine resulted in a notable decrease in the expression of several senescence-associated markers. One significant marker is β-galactosidase, an enzyme that, under normal physiological conditions, helps break down complex sugars into simpler sugars to provide energy. However, in the context of cellular aging, increased activity of β-galactosidase is often used as a biomarker for senescence. Berberine's effectiveness in reducing the activity of this enzyme suggests a decline in the senescent cell population, which is a positive indicator of its potential to mitigate aging processes at the cellular level.

Furthermore, Berberine also impacted the expression of p21, a cyclin-dependent kinase inhibitor that plays a crucial role in cell cycle regulation. p21 is typically upregulated in senescent cells, where it contributes to the halt in cellular division that defines senescence. The downregulation of p21 by Berberine points to its capacity to suppress the entry into or the maintenance of the senescent state in cells, further affirming its role in promoting cellular rejuvenation and reducing senescence-related changes.

The Potential of Berberine and DHB in Promoting Metabolic Health

Berberine and DHB are not only recognized for their anti-aging properties but also for their significant impact on metabolic health, particularly in glucose and lipid metabolism. These compounds have shown promising results in improving the body’s metabolic functions, with one of the most notable benefits being the regulation of blood glucose levels.

DHB, in particular, has been extensively studied for its ability to enhance the body's response to insulin, a critical hormone that regulates blood sugar levels. Insulin facilitates the uptake of glucose from the bloodstream into cells, providing them with essential energy. In healthy individuals, this process helps maintain blood glucose levels within a narrow range. However, in conditions like insulin resistance and type 2 diabetes, cells fail to respond adequately to insulin, leading to elevated blood sugar levels, which if left unchecked, can result in various health complications.

Berberine and DHB have been found to improve insulin sensitivity, meaning they help restore the cells’ responsiveness to insulin. This effect is particularly beneficial for individuals experiencing insulin resistance, as it can help lower high blood sugar levels and reduce the risk of diabetes-related complications. 

The mechanisms through which Berberine and DHB enhance insulin sensitivity involve several pathways, including the activation of AMPK. In 2008, Professor Nigel Turner at the Victor Chang Cardiac Research Institute released a pivotal study highlighting that DHB can significantly enhance insulin sensitivity by activating AMPK. This discovery has profound implications for treating metabolic disorders, particularly type 2 diabetes.

AMPK Activation: A Cornerstone for Metabolic Regulation

AMPK serves as a metabolic master switch, regulating several intracellular systems, primarily in response to changes in energy status. Activation of AMPK occurs under conditions of increased cellular AMP, which is an indicator of low energy levels. When a cell's energy is depleted, ATP (adenosine triphosphate) is converted to AMP (adenosine monophosphate). Unlike ATP, which stores energy with its three phosphate groups, AMP, with only one phosphate group, signifies reduced energy availability.

The rise in AMP levels triggers AMPK activation, which in turn initiates a cascade of responses aimed at restoring energy balance. AMPK activation inhibits the mTOR pathway, conserving energy by curbing cellular growth and proliferation. Simultaneously, it promotes the translocation of glucose transporters to the cell membrane, a process that increases glucose uptake from the bloodstream into the cells, effectively lowering blood sugar levels [10].

The Multifaceted Benefits of AMPK Activation by DHB

By activating AMPK, DHB facilitates several metabolic processes:

  • Enhanced Glucose Uptake: AMPK activation increases the number of glucose transporters on the cell surface, enhancing the uptake of glucose into the cells. This is particularly beneficial for cells that have become insulin resistant.

  • Reduced Hepatic Glucose Production: It decreases the production of glucose in the liver, directly contributing to lower blood glucose levels.

  • Improved Insulin Sensitivity: By enhancing glucose uptake and utilization, DHB helps improve the body’s response to insulin, making it more effective at lower concentrations.

Now let's turn the ways AMPK activation by DHB effects lipid metabolism.

The Impact of DHB on Cholesterol and Lipid Metabolism

Dihydroberberine (DHB) is gaining attention for its potential benefits in managing cholesterol levels, a key factor in cardiovascular health. Cholesterol is transported in our bloodstream by two main types of lipoproteins: high-density lipoprotein (HDL) and low-density lipoprotein (LDL).

To simplify their roles, imagine HDL as 'tow trucks' and LDL as 'delivery trucks' within the bloodstream. HDL, or 'good' cholesterol, acts like a tow truck by picking up excess cholesterol from the blood vessels and transporting it to the liver for removal. This process helps keep the arteries clear of buildup, reducing the risk of cardiovascular diseases.

Conversely, LDL, or 'bad' cholesterol, functions like delivery trucks. They distribute cholesterol throughout the body, but when there's too much LDL, it can deposit cholesterol in the artery walls, leading to clogged arteries, heart attacks, and strokes.

A 2022 study published in Nature highlighted DHB’s ability to favorably alter cholesterol levels. DHB was shown to lower LDL while increasing HDL levels, thereby improving the cholesterol profile. This beneficial effect is achieved partly by inhibiting the activity of acetyl-CoA carboxylase (ACC), a key enzyme in fatty acid synthesis.

ACC is involved in converting acetyl-CoA, a metabolic intermediate, into malonyl-CoA, the first step in fatty acid synthesis. These fatty acids are then used to form triglycerides, which are packaged with cholesterol into very low-density lipoproteins (VLDLs). As VLDLs circulate in the bloodstream, they deliver triglycerides to tissues, and in the process, they are converted into LDL particles [11].

By inhibiting ACC, DHB reduces the synthesis of fatty acids, thereby decreasing the liver's production of VLDLs. With fewer VLDLs released into circulation, the subsequent formation of LDL particles is also reduced. This action directly addresses the root cause of LDL buildup and helps improve the overall lipid profile. Additionally, by promoting a higher HDL level, DHB enhances the body’s ability to remove cholesterol from arteries, further protecting against atherosclerosis and associated cardiovascular diseases [11].

Role of DHB and Berberine in Modulating Rapamycin’s Effects

Rapamycin has been widely studied for its potential to extend healthspan and combat age-related diseases by specifically inhibiting the mTOR pathway, a critical regulator of cellular growth and metabolism. While promising, Rapamycin's use is sometimes associated with undesirable metabolic side effects, such as increased glucose and lipid levels, particularly when administered in high doses or on a chronic daily schedule.

Understanding mTOR Complexes and Their Functions

mTOR functions through two distinct complexes: mTORC1 and mTORC2. mTORC1 is well-known for its role in promoting cellular growth by controlling protein synthesis, nutrient sensing, and autophagy. Inhibition of mTORC1 is primarily targeted by Rapamycin and is beneficial for reducing the processes that drive aging and cancer.

On the other hand, mTORC2 is crucial for cell survival, regulating cell shape, lipid metabolism, and insulin signaling. This complex is essential for maintaining proper glucose homeostasis and lipid metabolism. mTORC2's role in insulin signaling makes it particularly important in managing metabolic health. Unlike mTORC1, mTORC2 is less sensitive to Rapamycin, especially at lower or intermittent doses. However, higher doses or continuous administration can inadvertently inhibit mTORC2, leading to disrupted insulin signaling and adverse metabolic effects such as hyperglycemia and hyperlipidemia.

It’s important to note, researchers found that these metabolic impairments caused by mTORC2 inhibition are reversible. When Rapamycin treatment is stopped, the markers of glucose homeostasis return to normal within 1-2 weeks, indicating that the adverse effects are due to the ongoing presence of the drug rather than permanent changes in the body's metabolic function [12].

AMPK: A Key Modulator in mTOR Signaling and Rapamycin Side Effects

AMPK is widely recognized for its role in energy balance and metabolic stress management within cells. A pivotal study published in 2019 in the journal Science demonstrated that AMPK does more than just inhibit mTORC1—it also directly activates mTORC2, highlighting its dual role in cellular regulation [13].

The study revealed that increasing AMPK activity, for example through the use of metformin, a common diabetes medication, leads to enhanced signaling through mTORC2. This finding is significant because mTORC2 plays a crucial role in cell survival, particularly under conditions of acute energetic stress. It is involved in critical processes such as lipid metabolism and the regulation of glucose uptake through insulin signaling.

Implications for Rapamycin Therapy

For patients using rapamycin, primarily for its healthspan-enhancing and anti-aging effects via the inhibition of mTORC1, the side effects of mTORC2 inhibition pose a challenge. These can include insulin resistance and disrupted glucose metabolism, which are substantial concerns given their implications for metabolic health. The inhibition of mTORC2 can undermine the benefits of rapamycin by compromising the body’s ability to manage glucose effectively, leading to broader metabolic disturbances.

However, the activation of mTORC2 by AMPK presents a potential strategy to mitigate these side effects. Agents that boost AMPK activity, such as DHB, berberine, or metformin, can play a critical role in balancing the effects of rapamycin therapy. By enhancing AMPK activity, these agents help promote mTORC2 activity even in the presence of rapamycin, thus supporting cellular functions that are vital for energy balance and metabolic health.

Using AMPK activators in conjunction with rapamycin could be a promising approach to reduce its metabolic side effects while still reaping the anti-aging benefits. This strategy leverages the unique ability of AMPK to simultaneously downregulate mTORC1 and upregulate mTORC2, optimizing therapeutic outcomes and minimizing risks. It represents a refined method to harness the benefits of rapamycin without the full extent of its drawbacks, particularly in the context of long-term use for healthspan enhancement.

Considerations and Future Directions

While the potential benefits of Dihydroberberine are noteworthy, there is still a gap in research. Many studies have been conducted on small sample sizes, and large-scale studies are needed to better ascertain the full spectrum of benefits and limitations of DHB use.

With regards to Rapamycin and DHB combined, there is a scarcity of trials evaluating the combined effects of these two drug candidates, with only the study by Guo et al. in 2014 testing both.

Moreover, most of the studies conducted so far have focused on Berberine rather than the chemically modified variant DHB. Comparative analyses of Berberine and DHB clearly demonstrate the superior potential of the latter in delivering therapeutic effects. Hence, studies assessing both Berberine and DHB need to be conducted to evaluate which form is most optimal for different types of treatments.

There is also a need for standardized preparation methods to ensure consistent potency and purity of Berberine and DHB supplements. Regulatory frameworks must oversee the quality of Berberine and DHB products. More large-scale, randomized controlled trials are necessary to confirm the efficacy and safety of Berberine and DHB in diverse populations, focusing on long-term outcomes, including C-reactive protein levels. C-reactive protein levels serve as a marker of inflammation. In the context of Berberine and DHB use, tracking the level of these proteins can help assess the agents' anti-inflammatory effects.

Further research into the precise molecular mechanisms of Berberine and DHB will also help optimize their use in clinical practice by understanding how they interact with various metabolic and signaling pathways, leading to targeted therapeutic strategies. As with many treatments, individual responses to Berberine and DHB can vary, so research into genetic markers that predict response to such agents could enable personalized treatment plans, maximizing efficacy and minimizing adverse effects.

Conclusion

DHB stands out as a promising derivative of Berberine, offering enhanced bioavailability and therapeutic efficacy. By chemically modifying Berberine to create DHB, researchers have significantly improved its absorption and potency, making it more effective at lower doses and reducing the risk of gastrointestinal side effects.

Berberine and DHB's roles also extend into the longevity and anti-aging sphere. By targeting pathways like mTOR and AMPK, these compounds help mitigate cellular senescence, oxidative stress, and metabolic dysfunctions. This is particularly significant in promoting healthy aging and potentially reducing age-related diseases.

Moreover, DHB and Berberine show potential in improving metabolic health, enhancing insulin sensitivity, and optimizing lipid profiles, thus offering benefits for conditions like diabetes and hyperlipidemia. Their ability to counteract the metabolic side effects of Rapamycin further underscores their therapeutic versatility. The synergistic effects of Berberine and Rapamycin in cancer treatment add another dimension to their potential, highlighting the need for further research into these compounds as complementary therapies.

Advancements in DHB research emphasize its potential as a powerful and versatile supplement, capable of addressing a broad spectrum of health issues while enhancing the beneficial properties of both Berberine and Rapamycin.

Citations

  1. Junio, H. A., Sy-Cordero, A. A., Ettefagh, K. A., Burns, J. T., Micko, K. T., Graf, T. N., ... & Cech, N. B. (2011). Synergy-directed fractionation of botanical medicines: a case study with goldenseal (Hydrastis canadensis). Journal of natural products, 74(7), 1621-1629.

  2. Li, Z., Zheng, J., Zhang, N., & Li, C. (2016). Berberine improves airway inflammation and inhibits NF-κB signaling pathway in an ovalbumin-induced rat model of asthma. Journal of Asthma, 53(10), 999–1005. https://doi.org/10.1080/02770903.2016.1180530

  3. Yin J, Xing H, Ye J. Efficacy of berberine in patients with type 2 diabetes mellitus. Metabolism. 2008 May;57(5):712-7. doi: 10.1016/j.metabol.2008.01.013. PMID: 18442638; PMCID: PMC2410097.

  4. Li, Z., Geng, Y. N., Jiang, J. D., & Kong, W. J. (2014). Antioxidant and anti-inflammatory activities of Berberine in the treatment of diabetes mellitus. Evidence-Based Complementary and Alternative Medicine, 2014.

  5. Kulkarni SK, Dhir A. Berberine: a plant alkaloid with therapeutic potential for central nervous system disorders. Phytother Res. 2010 Mar;24(3):317-24. doi: 10.1002/ptr.2968. PMID: 19998323.

  6. Moon JM, Ratliff KM, Hagele AM, Stecker RA, Mumford PW, Kerksick CM. Absorption Kinetics of Berberine and Dihydroberberine and Their Impact on Glycemia: A Randomized, Controlled, Crossover Pilot Trial. Nutrients. 2021 Dec 28;14(1):124. doi: 10.3390/nu14010124. PMID: 35010998; PMCID: PMC8746601.

  7. Buchanan, B., Meng, Q., Poulin, M. M., Zuccolo, J., Azike, C. G., Gabriele, J., & Baranowski, D. C. (2018). Comparative pharmacokinetics and safety assessment of transdermal Berberine and dihydroberberine. PloS one, 13(3), e0194979. https://doi.org/10.1371/journal.pone.0194979

  8. Nixon, R. A. (2013). The role of autophagy in neurodegenerative disease. Nature Medicine, 19(8), 983-997. DOI: 10.1038/nm.3232.

  9. Zhao H, Halicka HD, Li J, Darzynkiewicz Z. Berberine suppresses gero-conversion from cell cycle arrest to senescence. Aging (Albany NY). 2013 Aug;5(8):623-36. doi: 10.18632/aging.100593. PMID: 23974852; PMCID: PMC3796215.

  10. Nigel Turner, Jing-Ya Li, Alison Gosby, Sabrina W.C. To, Zhe Cheng, Hiroyuki Miyoshi, Makoto M. Taketo, Gregory J. Cooney, Edward W. Kraegen, David E. James, Li-Hong Hu, Jia Li, Ji-Ming Ye; Berberine and Its More Biologically Available Derivative, Dihydroberberine, Inhibit Mitochondrial Respiratory Complex I: A Mechanism for the Action of Berberine to Activate AMP-Activated Protein Kinase and Improve Insulin Action. Diabetes 1 May 2008; 57 (5): 1414–1418. https://doi.org/10.2337/db07-1552

  11. Ma, SR., Tong, Q., Lin, Y. et al. Berberine treats atherosclerosis via a vitamin-like effect down-regulating Choline-TMA-TMAO production pathway in gut microbiota. Sig Transduct Target Ther 7, 207 (2022). https://doi.org/10.1038/s41392-022-01027-6

  12. Salmon A. B. (2015). About-face on the metabolic side effects of rapamycin. Oncotarget, 6(5), 2585–2586. https://doi.org/10.18632/oncotarget.3354

  13. Kazyken D, Magnuson B, Bodur C, Acosta-Jaquez HA, Zhang D, Tong X, Barnes TM, Steinl GK, Patterson NE, Altheim CH, Sharma N, Inoki K, Cartee GD, Bridges D, Yin L, Riddle SM, Fingar DC. AMPK directly activates mTORC2 to promote cell survival during acute energetic stress. Sci Signal. 2019 Jun 11;12(585):eaav3249. doi: 10.1126/scisignal.aav3249. PMID: 31186373; PMCID: PMC6935248.

Dismiss

Stay Updated

Sign up for The Longevity Blueprint, a weekly newsletter from Healthspan analyzing the latest longevity research.

footer image