Urolithin A: The anti-aging secret of intestinal flora metabolites

Sep 08 2025

Urolithin A does not come directly from food; its formation relies on a complex biotransformation process. Its source is ellagitannins, which are widely found in plants such as pomegranates, walnuts, and berries. Upon entering the body, the ellagitannins are first hydrolyzed in the intestines, releasing ellagic acid. Specific strains of the intestinal flora, such as Gordonia and Lactobacillus, then play a key role in further metabolizing ellagic acid to produce urolithin A. This process fully demonstrates the interaction between the human intestinal microbiome and food components, and also reveals that the natural properties of urolithin A are highly dependent on the intestinal microbiome. Studies have found that only approximately 40% of the population are able to efficiently convert urolithin A, indicating that individual differences in intestinal microbiota significantly influence the synthesis of urolithin A.
Natural urolithin A cannot be directly absorbed from food; it must be synthesized through a complex and sophisticated intestinal metabolic chain: ellagitannins → ellagic acid → urolithin A. In this metabolic chain, a key, rate-limiting step is the ring-opening and hydroxylation of ellagic acid. This step acts as a bottleneck in the entire synthesis process, directly determining the ultimate efficiency of urolithin A production.

The diversity of the intestinal microbiome plays a crucial role in this process. A rich and diverse intestinal microbiome provides a wider variety of enzymes that catalyze the conversion of ellagic acid to urolithin A. An imbalance in the intestinal microbiome, such as with long-term antibiotic use, can significantly deplete beneficial bacteria, leading to a decrease in the number of strains critical for urolithin A synthesis and inhibiting its production. The intestinal pH also influences the synthesis process. A suitable pH environment maintains the activity of relevant enzymes and promotes the reaction. Excessively high or low pH can inhibit or even inactivate enzyme activity, hindering urolithin A synthesis. Host genetic factors also play a crucial role. Genetic background determines the initial composition and metabolic capacity of an individual’s gut microbiome. Some individuals may be predisposed to possess gut microbiota and metabolic genes that are more conducive to urolithin A synthesis, while others may be less so.

Research has shown that dietary structure significantly regulates the efficiency of urolithin A synthesis. A high-fiber diet provides abundant nutrients to beneficial gut microbiota, promoting the enrichment of urolithin A-producing bacterial genera such as Akkermansia and Bacteroides. Increased abundance of these genera effectively enhances urolithin A synthesis efficiency. Conversely, an unhealthy diet, such as excessive intake of high-fat and high-sugar foods, can disrupt the balance of the gut microbiome and inhibit urolithin A synthesis.

Core Biological Functions: Deep Regulation from Cells to Organs

Activation of Mitochondrial Autophagy: The “Scavenger” of the Cellular Energy Factory

Mitochondria are considered the “energy factories” of the cell, producing ATP through oxidative phosphorylation, providing energy for various cellular activities. However, as cellular metabolic activity continues, mitochondria inevitably become damaged and dysfunctional. These damaged mitochondria not only fail to efficiently produce energy but also produce large amounts of reactive oxygen species (ROS), which further damage other intracellular biomolecules such as DNA, proteins, and lipids, accelerating cell aging and death.
Urolithin A acts like a “scavenger,” specifically responsible for cleaning up these damaged mitochondria. It does so by targeting the mitophagy pathway, most critically the PINK1/Parkin signaling axis. When mitochondria are damaged, the mitochondrial membrane potential decreases, leading to the accumulation and activation of PINK1 on the outer mitochondrial membrane. Activated PINK1 recruits Parkin, a ubiquitin ligase that attaches ubiquitin molecules to mitochondrial membrane proteins. These ubiquitin-tagged mitochondria are then recognized and encapsidated by autophagosomes, forming autophagolysosomes, where they are ultimately degraded by hydrolytic enzymes within the lysosomes.

Urolithin A supplementation has demonstrated significant efficacy in the Caenorhabditis elegans model. It increased mitochondrial membrane potential by 30%, indicating a significant improvement in mitochondrial function, enabling more efficient energy conversion. ATP production increased by 25%, providing more energy for the nematodes’ vital activities. Muscle atrophy and decreased athletic ability, which are caused by mitochondrial dysfunction, were also delayed. This suggests that urolithin A, by activating mitophagy, effectively maintains mitochondrial quality control, safeguards cellular energy supply, and ultimately maintains normal physiological function. At a molecular level, urolithin A activates the expression of autophagy-related genes (ATG5 and ATG7). ATG5 and ATG7 are key proteins in the formation of autophagosomes, and their increased expression promotes autophagosome formation. Urolithin A also promotes the fusion of autophagosomes with lysosomes, enabling faster degradation and renewal of encapsulated mitochondria, maintaining a healthy mitochondrial population.

Anti-Inflammation and Oxidative Stress Regulation

Inflammation and oxidative stress are key pathological foundations for the development and progression of many diseases. They are interconnected, forming a vicious cycle that causes severe damage to the body. During the inflammatory response, immune cells are activated, releasing large amounts of pro-inflammatory cytokines, such as tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6). These pro-inflammatory cytokines further recruit immune cells, amplify the inflammatory response, and cause tissue damage. Oxidative stress is caused by an imbalance between the body’s oxidative and antioxidant systems. The excessive production of reactive oxygen species (ROS) overwhelms the body’s antioxidant defenses. ROS attack intracellular biomolecules, causing cell damage and apoptosis.

Urolithin A, a natural polyphenol derivative, plays an important role in anti-inflammatory and oxidative stress regulation. It reduces inflammation by inhibiting inflammatory pathways such as NF-κB and MAPK. NF-κB is an important transcription factor that plays a key role in inflammatory signaling. When cells are stimulated by inflammation, NF-κB is activated and translocated to the nucleus, initiating the transcription of a series of pro-inflammatory genes, leading to the massive release of pro-inflammatory cytokines. Urolithin A inhibits NF-κB activation and prevents its entry into the cell nucleus, thereby reducing the release of pro-inflammatory cytokines such as TNF-α and IL-6. Urolithin A also inhibits the MAPK signaling pathway, which includes multiple members such as ERK, JNK, and p38 MAPK. These pathways are activated during cellular stress and inflammation and are involved in regulating processes such as cell proliferation, differentiation, and apoptosis. By inhibiting the activation of these pathways, urolithin A reduces the transmission of inflammatory signals and alleviates the inflammatory response.

Urolithin A enhances the activity of antioxidant enzymes such as superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px). SOD catalyzes the conversion of superoxide anions to hydrogen peroxide, while GSH-Px reduces hydrogen peroxide to water. These two pathways work synergistically to effectively scavenge ROS in the body and reduce oxidative stress. Urolithin A reduces the levels of oxidative products such as malondialdehyde (MDA). MDA is a product of lipid peroxidation, and its level reflects the degree of oxidative damage in the body. Urolithin A’s reduced MDA production suggests that it can mitigate the cellular damage caused by lipid peroxidation.

Urolithin A’s effects are particularly pronounced in models of inflammatory bowel disease (IBD). IBD is a chronic, nonspecific inflammatory intestinal disease that includes ulcerative colitis and Crohn’s disease. Its pathogenesis is closely linked to intestinal inflammation and oxidative stress. Urolithin A can repair intestinal mucosal tight junction proteins (such as Claudin-1 and ZO-1). Claudin-1 and ZO-1 are important proteins that form tight junctions in the intestinal mucosa. Their normal expression and distribution are crucial for maintaining intestinal barrier function. In IBD patients, intestinal inflammation leads to damage of these tight junction proteins, increased intestinal permeability, and the translocation of endotoxins into the circulation, triggering systemic inflammation. Urolithin A can promote the expression and repair of Claudin-1 and ZO-1, reduce intestinal permeability by 40%, effectively prevent the translocation of endotoxins, relieve systemic inflammation, and improve the symptoms of IBD.

Muscle Protection and Improvement of Age-Related Muscle Atrophy

With aging, the body gradually loses muscle mass, leading to a decline in muscle strength and function. This phenomenon is known as age-related muscle atrophy (sarcopenia). This condition not only impacts the quality of life of older adults, increasing the risk of falls and fractures, but is also closely linked to the development and progression of many chronic diseases, such as cardiovascular disease and diabetes. Urolithin A demonstrates significant potential for muscle protection and improvement of age-related muscle atrophy.

Clinical studies have demonstrated that daily supplementation of 1g of urolithin A for four months in adults aged 65 and older increased handgrip strength and endurance by 7-8 times, demonstrating that urolithin A can significantly enhance muscle strength and endurance in older adults. The expression of mitochondrial biomarkers, such as PGC-1α, increased by 20% in leg muscles. PGC-1α is a key regulator of mitochondrial biogenesis. Increased expression indicates improved mitochondrial number and function, providing more energy to muscle cells and helping to maintain normal muscle function.

Urolithin A has multiple mechanisms of action. It not only clears damaged mitochondria and maintains energy metabolism in muscle cells, but also promotes muscle repair and regeneration by regulating the activity of muscle satellite cells. Muscle satellite cells are stem cells in muscle that possess the ability to self-renew and differentiate into myocytes, playing a crucial role in muscle repair and growth. Urolithin A activates muscle satellite cells, promoting their proliferation and differentiation, increasing the number and diameter of myofibers, and thereby improving muscle mass and strength. Urolithin A inhibits overactivation of the ubiquitin-proteasome system. In age-related muscle atrophy, the ubiquitin-proteasome system becomes overactivated, leading to increased muscle protein degradation and decreased muscle mass. Urolithin A inhibits the activity of key enzymes in this system, reducing muscle protein degradation and maintaining a balance between muscle protein synthesis and degradation, thereby slowing age-related muscle loss.

Systemic Modulation Effects: From Single Organ to Global Health

Potential Interventions for Neurodegenerative Diseases

Alzheimer’s disease and Parkinson’s disease are typical neurodegenerative diseases that severely impact patients’ quality of life and place a heavy burden on their families and society. Alzheimer’s disease is characterized by progressive cognitive impairment and memory loss. Patients develop amyloid-β deposits and tau hyperphosphorylation in the brain. These pathological changes lead to neuronal death and loss of synaptic function, ultimately causing cognitive and behavioral impairments. Parkinson’s disease, on the other hand, is characterized by motor symptoms such as bradykinesia, tremor, and muscle rigidity. Its pathological basis is the progressive degeneration of dopaminergic neurons in the substantia nigra-striatum pathway, resulting in decreased dopamine secretion.

Urolithin A has shown great potential for intervention in neurodegenerative diseases. Its ability to cross the blood-brain barrier (BBB) ​​is crucial for its neuroprotective effects. The BBB, a specialized barrier between blood and brain tissue, prevents many harmful substances from entering the brain, but also limits the therapeutic efficacy of many drugs. Urolithin A, due to its unique molecular structure and physicochemical properties, is able to successfully cross the BBB and enter the brain to exert its effects. In Alzheimer’s disease mice, urolithin A improved cognitive function and spatial memory by inhibiting Aβ amyloid deposition and Tau hyperphosphorylation. Aβ amyloid deposition leads to the formation of senile plaques, while Tau hyperphosphorylation causes neurofibrillary tangles, both of which are hallmark pathological features of Alzheimer’s disease. Urolithin A inhibits the misfolding and aggregation of these proteins, reducing the formation of senile plaques and neurofibrillary tangles, thereby protecting neurons and improving cognitive function. The underlying mechanism involves enhanced SIRT1 deacetylase activity and regulation of NAD+ metabolism. SIRT1 is an NAD+-dependent deacetylase that plays an important role in cellular metabolism, aging, and stress responses. Urolithin A activates SIRT1, enhancing its deacetylase activity, thereby regulating the functions of downstream proteins and reducing Aβ amyloid deposition and Tau hyperphosphorylation. Urolithin A also regulates NAD+ metabolism, increasing intracellular NAD+ levels and providing sufficient substrate for SIRT1 activation, further enhancing its neuroprotective effects.

In Parkinson’s disease models, urolithin A protects dopaminergic neuronal mitochondrial function, reduces α-synuclein aggregation, and slows degeneration of the substantia nigra-striatum pathway. Mitochondrial dysfunction and α-synuclein aggregation are key components of Parkinson’s disease pathogenesis. Urolithin A activates mitophagy, clearing damaged mitochondria and maintaining normal mitochondrial function, thereby providing sufficient energy for dopaminergic neurons. Urolithin A also inhibits α-synuclein aggregation, reducing its toxic effects on neurons, thereby protecting dopaminergic neurons and slowing degeneration of the substantia nigra-striatum pathway.

Metabolic Syndrome and Cardiovascular Protection

Metabolic syndrome is a cluster of metabolic disorders characterized by obesity, hyperglycemia, dyslipidemia, and hypertension. It is closely associated with the development and progression of cardiovascular disease. Obesity is a key component of metabolic syndrome. Excessive fat accumulation can lead to insulin resistance and increased inflammation, which in turn can cause problems such as dysglycemia, dyslipidemia, and hypertension. These metabolic disorders can further damage vascular endothelial cells, promote the development of atherosclerosis, and increase the risk of cardiovascular disease.
Urolithin A plays a key role in metabolic syndrome and cardiovascular protection. It promotes glucose uptake by activating the AMPK pathway, lowering fasting blood glucose by 15%-20%. AMPK is a cellular energy sensor that becomes activated when cellular energy levels decrease, regulating metabolic processes within cells, promoting glucose uptake and utilization, improving insulin sensitivity, and thus lowering blood glucose levels. Urolithin A also inhibits hepatic HMG-CoA reductase, reducing LDL-C synthesis. HMG-CoA reductase is a key enzyme in cholesterol synthesis. Urolithin A inhibits this enzyme, reducing cholesterol synthesis and lowering LDL-C levels in the blood, thereby reducing the risk of atherosclerosis.

Urolithin A improves endothelial function in atherosclerosis models, promotes NO release, and inhibits platelet aggregation, thereby reducing the risk of plaque formation. Endothelial cells, the cells lining the blood vessels, secrete substances such as nitric oxide (NO), regulating vasodilation and contraction, inhibiting platelet aggregation, and inflammatory responses. When endothelial function is impaired, NO secretion decreases, platelet aggregation increases, and inflammation increases, all of which promote the formation of atherosclerotic plaques. Urolithin A protects endothelial cells, promotes NO release, enhances vasodilation, and inhibits platelet aggregation, thereby reducing the risk of atherosclerotic plaque formation.

Urolithin A accelerates fatty acid oxidation by inducing browning of white fat and activation of brown fat, thereby alleviating diet-induced obesity. White fat primarily stores energy, while brown fat consumes energy through heat production. Urolithin A can induce the conversion of white adipocytes into brown adipocytes, increasing the content of brown fat. It also activates brown adipocytes, enhancing their heat production and accelerating fatty acid oxidation, thereby reducing fat accumulation and alleviating obesity.

Intestinal Barrier and Maintaining Immune Homeostasis

The intestine is the body’s largest immune organ, and intestinal barrier function and immune homeostasis are crucial for maintaining human health. The intestinal barrier, composed of physical, chemical, biological, and immune barriers, prevents the invasion of pathogens and harmful substances, maintaining a stable intestinal environment. Immune homeostasis refers to the immune system’s ability to mount an appropriate immune response to pathogens while preventing excessive immune responses from causing damage to the body.

Urolithin A, a metabolite of the intestinal flora, negatively regulates the intestinal microbiome, promoting mucus secretion and antimicrobial peptide expression, thereby enhancing intestinal mucosal barrier function. The intestinal microbiome refers to the microbial community within the intestine and is closely related to human health. Urolithin A can regulate the composition and function of the intestinal flora, promoting the growth of beneficial bacteria and inhibiting the proliferation of harmful bacteria, thereby maintaining a balanced intestinal microbiome. Urolithin A also promotes the secretion of the intestinal mucus layer, which forms a physical barrier to prevent pathogen invasion. Urolithin A also promotes the expression of antimicrobial peptides, which have antibacterial and antiviral properties and can enhance the intestinal immune defenses.

In a model of ulcerative colitis, urolithin A restored tight junction protein expression by 60% and reduced inflammation scores by 35%. Ulcerative colitis is a chronic, nonspecific intestinal inflammatory disease whose pathogenesis is closely related to impaired intestinal barrier function and imbalanced immune homeostasis. In patients with ulcerative colitis, intestinal tight junction proteins are damaged, intestinal permeability is increased, and endotoxins are translocated into the bloodstream, triggering systemic inflammation. Urolithin A promotes the expression and repair of tight junction proteins, reducing intestinal permeability, effectively preventing the translocation of endotoxins, alleviating systemic inflammation, and improving the symptoms of ulcerative colitis. Urolithin A also shows therapeutic potential for chronic inflammatory diseases such as Crohn’s disease by regulating the Th17/Treg cell balance and inhibiting excessive immune responses. Th17 cells and Treg cells are two important immune cells that play opposing roles in immune regulation. Th17 cells secrete proinflammatory cytokines and promote inflammatory responses, while Treg cells suppress immune responses and maintain immune homeostasis. In chronic inflammatory diseases, the Th17/Treg cell balance is disrupted, with Th17 cells becoming hyperactive and Treg cells becoming deficient, leading to excessive immune responses. Urolithin A can regulate the Th17/Treg cell balance, inhibiting Th17 cell function and enhancing Treg cell function, thereby suppressing excessive immune responses and alleviating the symptoms of chronic inflammatory diseases.

 

Application Areas and Research Progress

Development of Functional Foods and Health Products

Urolithin A has already made a name for itself in the functional food and health product sector. Dietary supplements with UA as their core ingredient have successfully entered the market, boasting benefits such as anti-aging, muscle building, and improved sleep, attracting significant consumer interest. Common dosage forms include enteric-coated capsules, typically containing 500mg per capsule. This ensures effective intestinal absorption of UA, preventing damage from stomach acid. Sublingual tablets are also a common dosage form, offering the advantage of direct absorption through the sublingual mucosa, rapid entry into the bloodstream, and enhanced bioavailability.

Multiple clinical trials have strongly supported the efficacy of these products. Studies in middle-aged and elderly individuals have shown that long-term use of urolithin A dietary supplements can significantly improve muscle endurance. In a four-month trial, participants supplemented with 1g of urolithin A daily and showed a 7-8-fold increase in hand grip strength and endurance. This has important implications for improving daily living abilities in older adults, enabling them to more easily perform tasks like holding a pen and lifting objects. Urolithin A also significantly improves sleep. It regulates the body’s circadian rhythm. For those experiencing fragmented sleep, long-term urolithin A supplementation significantly improved sleep quality, with fewer nighttime awakenings and more consistent, deep sleep. Studies have shown that its mechanism of action involves regulating the expression of clock genes such as BMAL1. By modulating the expression of these genes, urolithin A can regularize the body’s circadian clock, thereby improving sleep quality.

Frontier Explorations in the Medical Field

In the medical field, research on urolithin A is continuing to deepen, demonstrating significant potential. A Phase II clinical trial for Alzheimer’s disease is currently underway, evaluating the efficacy of UA in treating mild cognitive impairment. Alzheimer’s disease is a severe neurodegenerative disorder with no effective cure. Early intervention is crucial for slowing disease progression. Urolithin A, with its unique neuroprotective properties, holds promise as a new treatment for Alzheimer’s disease. Preclinical studies have shown that urolithin A can cross the blood-brain barrier and, once in the brain, inhibit Aβ amyloid deposition and Tau hyperphosphorylation, two key pathological changes characteristic of Alzheimer’s disease. Urolithin A’s inhibitory effects on these pathways offer the potential for improving cognitive function in patients.

Urolithin A has also made significant progress in the treatment of inflammatory bowel disease. Synthetic analogs, due to improved stability, have entered the efficacy validation phase in animal models. Inflammatory bowel disease (IBD) is a group of chronic inflammatory bowel diseases, including ulcerative colitis and Crohn’s disease. Current treatments have numerous limitations and are unable to effectively repair damaged intestinal barriers. Urolithin A and its synthetic analogs offer potential solutions to this problem. Research has shown that they can restore intestinal barrier integrity by increasing the production of proteins that strengthen intestinal epithelial cell junctions and reducing intestinal inflammation. In animal studies, mice treated with synthetic analogs of urolithin A showed significant reductions in intestinal inflammation, restored expression of intestinal mucosal tight junction proteins (such as claudin-1 and ZO-1), decreased intestinal permeability, effectively prevented endotoxin translocation, and alleviated systemic inflammation.

In addition to the aforementioned areas, research on urolithin A in adjuvant cancer therapy and osteoporosis prevention is also underway. In the adjuvant cancer therapy setting, studies have shown that urolithin A can inhibit angiogenesis and cancer cell proliferation. Tumor growth and metastasis depend on the formation of new blood vessels. Urolithin A reduces tumor angiogenesis by inhibiting signaling pathways such as vascular endothelial growth factor (VEGF), thereby limiting tumor nutrient supply and inhibiting growth and metastasis. Urolithin A can also induce apoptosis in cancer cells, activating intracellular apoptotic signaling pathways and promoting programmed cell death. In the prevention of osteoporosis, urolithin A can promote osteoblast differentiation and increase bone density. Osteoblasts are the cells responsible for bone formation. Urolithin A can regulate the expression of osteoblast-related genes, promote their proliferation and differentiation, and enhance bone matrix synthesis and mineralization, thereby preventing the development of osteoporosis.

Cosmetic and Skin Health Applications

Leveraging its potent antioxidant and mitochondrial protective properties, urolithin A has also been widely used in the cosmetic and skin health fields. It is incorporated into various anti-aging skincare products, becoming a new favorite in the skincare industry. Urolithin A is highly effective in combating photoaging. Ultraviolet light is a major factor in photoaging, inducing the production of large amounts of reactive oxygen species (ROS), which damage collagen and elastin fibers in the skin, leading to skin sagging, increased wrinkling, and hyperpigmentation. Urolithin A can effectively mitigate UV-induced photoaging damage by scavenging ROS within skin cells, reducing oxidative stress and protecting them from damage.

Urolithin A also promotes collagen synthesis, improving skin elasticity and radiance. In experiments with skin fibroblasts, a 0.5% UA preparation reduced the expression of β-galactosidase, a marker of fibroblast senescence, by 25%, demonstrating that urolithin A can slow the aging process of skin cells. Mechanistically, urolithin A activates the transforming growth factor-β (TGF-β)/Smad pathway, upregulating the expression of collagen genes (such as COL1A1 and COL3A1) and increasing collagen synthesis. Urolithin A inhibits the activity of matrix metalloproteinases (MMPs), particularly MMP-1, MMP-3, and MMP-9. These enzymes degrade collagen and other extracellular matrix components during skin aging. Urolithin A’s inhibitory effect reduces collagen degradation, effectively reducing wrinkles, improving skin elasticity, and making the skin firmer and more radiant, demonstrating a unique “endogenous anti-aging” advantage.

 

Safety and Future Outlook

Safety Assessment and Individual Differences

In terms of safety, existing toxicological data strongly support the use of urolithin A. Acute toxicity studies in rats showed that the median lethal dose (LD₅₀) of urolithin A was >2000 mg/kg, indicating that at higher doses, urolithin A exhibits low acute toxicity and is relatively safe. Long-term ingestion studies also demonstrated that rats exposed to urolithin A at a dose of 100 mg/kg/day did not experience significant liver or kidney damage. However, individual differences cannot be ignored in the use of urolithin A. Because the bioavailability of urolithin A is highly dependent on the intestinal microbiota, individual differences in intestinal microbiome composition and function lead to variations in the metabolism and effects of urolithin A.

Some individuals with intestinal sensitivities may experience transient intestinal discomfort, such as mild diarrhea and abdominal distension, when supplementing with urolithin A. This may be because urolithin A, in its process of regulating the intestinal microbiome, stimulates the intestinal microbial community and intestinal mucosa. To implement personalized supplementation plans, genetic testing and microbiome analysis are particularly important. Genetic testing can reveal an individual’s genetic background and analyze genetic polymorphisms related to urolithin A metabolism, thereby predicting an individual’s metabolic capacity and response to urolithin A. Microbiome analysis can provide a direct understanding of the composition and function of an individual’s intestinal microbiome, determining whether it has the ability to efficiently convert and produce urolithin A. For individuals whose intestinal microbiome composition is not conducive to urolithin A synthesis, dietary adjustments and probiotic supplementation can be used to improve the intestinal microbiome and enhance the synthesis and utilization efficiency of urolithin A.

Technological Breakthroughs and Industry Directions

Synthetic Biology Breakthroughs: Scientists have achieved significant breakthroughs in synthetic biology, providing a new approach for the large-scale production of urolithin A. By engineering Escherichia coli and constructing a multi-enzyme cascade reaction system, they have successfully achieved efficient biosynthesis of urolithin A. In this system, multiple enzymes work synergistically to mimic the biosynthesis pathway of urolithin A in nature, but with greater efficiency and controllability. The optimized production process has enabled a yield of urolithin A of 15g/L, a significant increase compared to traditional methods, paving the way for its large-scale application. The purity of the product has also been significantly improved, reaching >99%. This high-purity urolithin A can better meet the stringent raw material quality requirements of pharmaceutical and health product applications, reducing potential risks from impurities. This breakthrough effectively addresses the high cost and low conversion rates associated with natural extraction methods, significantly reducing the production cost of urolithin A and significantly improving production efficiency, providing strong support for its widespread market promotion and application.

Delivery System Innovation: To enhance the water solubility and targeting of urolithin A, researchers have innovated the delivery system, developing nanoliposome and microcapsule formulations. Nanoliposomes are tiny particles composed of a phospholipid bilayer, ranging in size from 1 to 100 nanometers, that can encapsulate urolithin A. Due to their excellent solubility, stability, and biocompatibility, nanoliposomes are able to traverse capillary walls and enter target tissues during blood circulation, thereby enhancing the targeting of urolithin A and enabling it to act more precisely on target cells and tissues. Nanoliposomes also protect urolithin A from degradation by enzymes and other substances in the body, prolonging its duration of action. Microencapsulated formulations encapsulate urolithin A within tiny capsules. By controlling the release mechanism of the capsules, urolithin A is slowly released, improving its bioavailability. These novel delivery systems are expected to increase the oral bioavailability of urolithin A from 30% to over 60%, significantly improving its efficacy, reducing its dosage, and reducing potential side effects.

Precision Medicine Applications: With the deepening of research on urolithin A, its application prospects in precision medicine are expanding. Combining gut microbiome analysis with metabolomics analysis can develop personalized urolithin A supplementation regimens. Gut microbiome testing can reveal the composition and function of an individual’s gut microbiome and assess its ability to synthesize and metabolize urolithin A. Metabolomics analysis can detect changes in metabolites within an individual’s body and assess the impact of urolithin A on their metabolism. By comprehensively analyzing this information, physicians can tailor the dosage and duration of urolithin A supplementation for their patients, achieving precision medicine. For patients with metabolic syndrome, physicians can determine the most appropriate urolithin A supplementation regimen based on their gut microbiome and metabolomics profile to achieve optimal therapeutic outcomes. This precision medicine approach is driving the transformation of urolithin A from a functional ingredient to a precision anti-aging drug, providing new insights and approaches for future health management and disease prevention.

Research on urolithin A has revealed the profound connection between gut microbiome metabolites and human health. Its transformation from a “natural product” to a “systemic modulator” marks a paradigm shift in anti-aging research from a single target to holistic regulation. With the accumulation of synthetic technology and clinical evidence, this “natural molecule” derived from the synergy of plants and microorganisms is expected to become a core intervention for future health management and disease prevention.

Online Consultation
Return Top