GENEMEDICS APP
GENEMEDICS NUTRITION
GW0742 is a selective peroxisomeproliferator-activated receptor (PPAR)-Ī“ agonist, which is produced mainly in the liver and plays a key role in the breakdown of fatty acids. It is almost similar in structure to Cardarine but GW0742 is 200 times more potent and has more health benefits.
As a PPAR delta agonist, GW0742 enhances fatty acid metabolism. This in turn preserves blood sugar as well as physical endurance. It is because of this effect that athletes, bodybuilders, and other physically active individuals use this PPAR delta agonist. Even in very small doses, GW0742 can significantly boost energy levels and stamina.
Studies suggest that GW0742 can help increase exercise endurance by promoting muscle regeneration:
GW0742 is a selective PPARĪ“ (Peroxisome Proliferator-Activated Receptor Delta) agonist that has been studied for its potential metabolic benefits, including fat loss and improved energy expenditure. By activating PPARĪ“, GW0742 enhances fatty acid oxidation, increases endurance, and promotes lean muscle retention, making it appealing for individuals looking to lose weight while preserving muscle mass. Additionally, it may improve insulin sensitivity and reduce inflammation, both of which are crucial for effective weight management. While research is still ongoing, GW0742 shows promise as a compound that supports fat loss and metabolic health, particularly when combined with a proper diet and exercise regimen.
By breaking down fatty acids, GW0742 improves body composition and helps lose weight:
GW0742, a potent PPAR-Ī“ agonist, has been studied for its potential cardiovascular benefits, particularly in reducing the risk of heart disease. By activating PPAR-Ī“, GW0742 enhances lipid metabolism, increases HDL (good cholesterol), and reduces LDL (bad cholesterol), which helps prevent atherosclerosis and arterial plaque buildup. Additionally, it improves endothelial function, reduces inflammation, and enhances mitochondrial efficiency in cardiac tissues, all of which contribute to better heart health. These effects collectively support improved circulation, reduced oxidative stress, and a lower risk of cardiovascular events such as heart attacks and strokes. While research is still ongoing, GW0742 shows promise as a therapeutic option for heart disease prevention and overall cardiovascular protection.
The ability of GW0742 to break down fatty acids produces beneficial effects on the heart, according to studies:
GW0742, a selective PPAR-Ī“ agonist, has been shown to accelerate wound healing by promoting cellular regeneration, reducing inflammation, and enhancing angiogenesis. By activating PPAR-Ī“ pathways, GW0742 stimulates keratinocyte proliferation and migration, which are essential for skin repair. It also boosts collagen synthesis and fibroblast activity, strengthening the extracellular matrix and improving tissue integrity. Additionally, GW0742 enhances blood vessel formation, ensuring better oxygen and nutrient delivery to the wound site. Its anti-inflammatory properties help reduce excessive scarring and promote a more efficient healing process. These combined effects make GW0742 a promising candidate for improving wound healing, particularly in conditions where healing is impaired, such as diabetes or chronic ulcers.
The regenerative properties of GW0742 are also backed by a number of studies:
GW0742, a potent PPAR-Ī“ agonist, has been shown to boost brain power by enhancing cognitive function, protecting neurons, and promoting neuroplasticity. By activating PPAR-Ī“ pathways, GW0742 supports mitochondrial function and energy metabolism in brain cells, leading to improved focus, memory, and mental clarity. Its neuroprotective effects help reduce oxidative stress and inflammation, which are linked to neurodegenerative diseases like Alzheimer’s and Parkinson’s. Additionally, GW0742 may enhance synaptic plasticity and stimulate the growth of new neurons, contributing to long-term brain health. These benefits make GW0742 a promising compound for improving cognitive performance and potentially delaying age-related cognitive decline.
Evidence suggests that GW0742 is integral for brain health:
GW0742, a selective PPAR-Ī“ agonist, has been shown to lower blood pressure by improving vascular function and reducing inflammation. By activating PPAR-Ī“ pathways, GW0742 enhances nitric oxide production, which promotes vasodilation and improves blood flow. This helps relax blood vessels, reducing resistance and lowering overall blood pressure levels. Additionally, GW0742 has been found to decrease oxidative stress and inflammation in the cardiovascular system, further supporting heart health. Its ability to enhance lipid metabolism and reduce arterial stiffness makes it a promising therapeutic candidate for managing hypertension and related cardiovascular conditions.
GW0742 can also lower blood pressure which can be beneficial for the treatment of hypertension:
GW0742, a potent PPAR-Ī“ agonist, has been shown to improve blood sugar levels and alleviate symptoms of diabetes by enhancing insulin sensitivity and glucose metabolism. By activating PPAR-Ī“, GW0742 promotes the uptake of glucose into muscle cells, reducing blood sugar spikes and improving overall glycemic control. Additionally, it helps regulate lipid metabolism, preventing the accumulation of excess fats that contribute to insulin resistance. Studies suggest that GW0742 also reduces inflammation and oxidative stress, two key factors in the progression of diabetes. These effects make GW0742 a promising candidate for managing diabetes and its complications, potentially offering an alternative approach to traditional treatments.
Studies show that GW0742 can help stabilize blood sugar levels and can be considered an anti-diabetic agent:
GW0742 has shown promising potential in treating lung injuries by reducing inflammation, oxidative stress, and tissue damage through activation of the PPAR-Ī“ pathway. This compound helps modulate immune responses in the lungs, limiting the production of pro-inflammatory cytokines that can worsen injury or lead to chronic conditions like fibrosis. In experimental models, GW0742 has been observed to improve oxygen exchange, preserve lung structure, and accelerate healing in damaged pulmonary tissues. These protective effects make it a compelling therapeutic candidate for conditions such as acute lung injury (ALI), acute respiratory distress syndrome (ARDS), and other inflammatory lung diseases.
A good deal of evidence shows that GW0742 can help protect the lungs against various forms of injury:
GW0742 may improve eye health by activating PPAR-Ī“ receptors, which play a role in reducing inflammation and protecting retinal cells from oxidative stress. This action can help preserve vision and may offer therapeutic benefits in conditions like diabetic retinopathy and age-related macular degeneration. While more human studies are needed, early research suggests that GW0742 supports ocular health by enhancing cellular energy metabolism and reducing damage to eye tissues.
The activation of PPARβ/Γ by GW0742 produces beneficial effects on visual function, according to studies:
GW0742 side effects are very uncommon. There have been some side effects associated with the use of this drug wherein the patient had one of the issues listed below at some point while being on GW0742. However, these side effects werenāt confirmed to be associated with the treatment and could have been a coincidence and not related to the use of GW0742. Despite this, it was listed as a side effect associated with GW0742 even though these associated side effects are very uncommon.
Side effects associated with GW0742 may include the following:
GW0742 and GW501516 (commonly known as Cardarine) are both PPAR-Ī“ agonists, but they have distinct characteristics and applications. GW501516 is well-known for its ability to enhance endurance, fat metabolism, and cardiovascular performance, making it popular among athletes and bodybuilders. It primarily stimulates fatty acid oxidation and mitochondrial biogenesis, leading to improved energy efficiency and increased stamina. However, concerns about potential carcinogenic effects in animal studies have raised safety questions regarding long-term use.
On the other hand, GW0742 is a more selective and potent PPAR-Ī“ agonist, with additional benefits beyond endurance and fat metabolism. It has been studied for its potential role in improving cardiovascular health, lowering blood pressure, and reducing inflammation, making it a promising candidate for treating metabolic disorders and chronic inflammatory diseases. GW0742 also appears to have neuroprotective and wound-healing properties, which distinguish it from GW501516.
While both compounds share similar mechanisms of action, GW0742 is considered to have a broader therapeutic scope with potential medical applications, whereas GW501516 is primarily used for performance enhancement. The safety profile of GW0742 is still under investigation, but its effects on cardiovascular and metabolic health may make it a more viable option for clinical use in the future.
The optimal dosage of GW0742 is not well-established for human use, as most studies have been conducted in animal models. In research settings, doses typically range from 1 to 10 mg/kg per day, depending on the specific condition being studied. However, due to its high potency as a PPAR-Ī“ agonist, lower doses may still provide significant metabolic and anti-inflammatory benefits.
Since GW0742 is not approved for human consumption, there are no standardized dosage guidelines. Users who experiment with GW0742 often start with lower doses, around 2-5 mg per day, to assess tolerance and potential effects. Higher doses may amplify benefits such as improved endurance, cardiovascular health, and fat metabolism, but they could also increase the risk of adverse effects.
Long-term safety data on GW0742 are limited, so careful monitoring of health markers is essential when using it. As with any research compound, it is crucial to consult healthcare professionals before experimenting with GW0742, especially for individuals with pre-existing medical conditions or those using other medications.
GW0742 has gained attention in the bodybuilding community due to its potential to enhance endurance, fat metabolism, and muscle recovery. As a potent PPAR-Ī“ agonist, it activates pathways that promote energy expenditure and reduce inflammation, making it appealing for athletes looking to improve overall performance. Its effects are often compared to GW501516 (Cardarine), though GW0742 is believed to have a stronger affinity for PPAR-Ī“ receptors.
One of the key benefits of GW0742 in bodybuilding is its ability to increase fatty acid oxidation, which can help users achieve a leaner physique while maintaining muscle mass. Additionally, its anti-inflammatory properties may support faster recovery, reducing muscle soreness and allowing for more intense training sessions. Some research also suggests that it can improve cardiovascular function, which is essential for endurance athletes and bodybuilders who engage in high-intensity workouts.
Despite its promising effects, GW0742 remains an experimental compound with limited human studies. Bodybuilders considering its use should be aware of potential risks, including unknown long-term side effects. Since it is not approved for human consumption, responsible use, proper dosing, and post-cycle health monitoring are crucial to minimizing risks while maximizing benefits.
GW0742, a potent PPAR-Ī“ agonist, is known for its relatively short half-life compared to some other compounds in its class. The estimated half-life of GW0742 is typically between 4 to 6 hours, meaning it is metabolized and cleared from the body relatively quickly. Because of this short duration, maintaining consistent plasma levels may require more frequent dosing or sustained-release formulations for therapeutic applications.
The half-life of GW0742 directly impacts how it is administered in both research and potential clinical settings. Frequent dosing may be necessary to ensure that the compound maintains its effects on metabolic regulation, inflammation reduction, or cardiovascular support throughout the day. This is especially important in experimental models where the timing and consistency of exposure influence results.
While the short half-life can be a limitation, it may also offer advantages, such as more precise control over the duration of its effects and potentially reduced risk of long-term accumulation or side effects. Researchers are exploring ways to optimize dosing strategies to maximize GW0742ās therapeutic potential while balancing safety and efficacy.
A typical GW0742 cycle for research purposes usually spans between 4 to 8 weeks, depending on the desired outcomes and the specific goals of the study. Researchers often administer the compound daily due to its short half-life, ensuring consistent activation of PPAR-Ī“ receptors throughout the cycle. Dosages are carefully measured, commonly ranging from 2 to 10 mg per day in animal models, although exact amounts vary based on body weight and experimental design.
During the GW0742 cycle, researchers may observe enhanced endurance, improved glucose metabolism, reduced inflammation, and potential fat oxidation effects. These outcomes are often monitored using biomarkers, performance tests, or imaging studies. Since GW0742 is non-steroidal, it does not typically require post-cycle therapy (PCT), but researchers still assess for hormonal or metabolic changes during and after the cycle to ensure safety.
At the end of a GW0742 cycle, itās important to evaluate recovery and long-term effects. Researchers often implement a washout period to allow the compound to clear from the system, monitoring for any residual effects or changes in physiological markers. This helps in understanding the full impact of GW0742 and optimizing future cycles for therapeutic or performance-based applications.
GW0742, while promising in preclinical research, has shown potential side effects that researchers closely monitor. One of the primary concerns is gastrointestinal discomfort, which may include nausea or mild cramping in some test subjects. These effects are generally dose-dependent and tend to lessen when lower dosages are used or when administered with food.
Another possible side effect observed in some studies is altered liver enzyme levels, indicating that GW0742 might exert some hepatic stress over extended use. Although itās not classified as hepatotoxic, regular liver function monitoring is recommended during research cycles, especially in long-term studies. Additionally, changes in lipid profiles, such as reduced HDL cholesterol, have also been noted.
Lastly, as GW0742 is a potent PPAR-Ī“ agonist, it can impact gene expression related to metabolism, inflammation, and cell growth. While this modulation is often the basis for its therapeutic potential, it may also carry unintended effects like hormonal imbalances or metabolic shifts if not carefully managed. More human research is needed to fully understand the long-term safety and side effect profile of GW074.
GW0742 is primarily used in scientific research to explore its role as a selective PPAR-Ī“ (peroxisome proliferator-activated receptor delta) agonist. This compound has shown potential in regulating lipid metabolism, enhancing endurance, and improving energy utilization, making it a promising candidate for studies on metabolic disorders and cardiovascular health. Researchers use GW0742 to understand how PPAR-Ī“ activation can influence fat burning and energy balance at the cellular level.
Another key use of GW0742 is in studies related to inflammation and immune response. Its anti-inflammatory properties have made it valuable in preclinical models of conditions like atherosclerosis, lung injury, and kidney inflammation. By targeting PPAR-Ī“, GW0742 helps modulate cytokine production and reduce oxidative stress, which are critical components in chronic inflammatory diseases.
GW0742 is also being explored for its potential benefits in treating diabetes and improving insulin sensitivity. Research indicates that it may help regulate blood glucose levels and improve lipid profiles in diabetic models. This has sparked interest in its use as a potential therapeutic for type 2 diabetes and metabolic syndrome, although further studies are needed to fully understand its safety and efficacy in humans.
This study shows that activation of PPAR-β/Ī“ promotes, while its inhibition reduces, preretinal neovascularization (NV) in models of ocular disease. In human retinal endothelial cells, the PPAR-β/Ī“ agonist GW0742āa selective small molecule agonistāincreased Angptl4 expression and enhanced tube formation, while the antagonist GSK0660 suppressed both proliferation and tube formation. In a rat model of oxygen-induced retinopathy, GW0742 elevated retinal Angptl4 levels and worsened NV, whereas GSK0660 significantly reduced NV. It activates the PPAR-Ī“ receptor, influencing metabolism and inflammation. These findings suggest that PPAR-β/Ī“ contributes to pathological ocular NV and that its inhibition may offer a novel therapeutic strategy.
Itās used in research to study metabolic diseases, cardiovascular health, and inflammation, with growing interest in its role in hepatic metabolism.
Its effects may last up to 24 hours, but human data is limited for this selective peroxisome proliferator.
Studies show improved lipid metabolism, reduced inflammation, and better endurance, which may be linked to its chemical structure.
It selectively activates the PPAR-Ī“ receptor to regulate gene expression related to energy use and can also influence insulin levels.
Itās also known as GSK3787 in some research contexts and has been shown to interact with key enzymes involved in metabolic regulation.
Injectable cardarine has an estimated half-life of about 20 to 24 hours, maintaining biological activity throughout that period.
Itās a research chemical name assigned to a synthetic PPAR-Ī“ agonist that influences the expression of target genes involved in energy regulation and lipid metabolism. Scientists study its modulation of target genes to explore potential therapeutic applications.
Potential effects include enhanced fat burning, reduced inflammation, and improved endurance.
It modulates energy metabolism and inflammatory pathways through PPAR-Ī“ activation.
Its primary use is in preclinical research for treating metabolic and inflammatory conditions.
Le Garf S, Murdaca J, Mothe-Satney I, et al. Complementary Immunometabolic Effects of Exercise and PPARβ/Γ Agonist in the Context of Diet-Induced Weight Loss in Obese Female Mice. Int J Mol Sci. 2019;20(20):5182. Published 2019 Oct 19. doi:10.3390/ijms20205182.
Complementary Immunometabolic Effects of Exercise and PPARβ/Γ Agonist in the Context of Diet-Induced Weight Loss in Obese Female Mice
This study shows that both aerobic exercise and treatment with GW0742, a PPARβ/Ī“ agonist, improve metabolic and anti-inflammatory responses in diet-induced obese female mice, especially when combined. Exercise increased AMPK activity, while GW0742 enhanced fatty acid oxidation in immune tissues, and together they boosted anti-inflammatory Foxp3+ T cells, reduced inflammation and fat mass, improved muscle oxidative capacity, and enhanced insulin sensitivityāsupporting PPARβ/Ī“ activation as a promising strategy for treating obesity-related metabolic dysfunction.
You can read the full article at https://pmc.ncbi.nlm.nih.gov/articles/PMC6829333/.
Mothe-Satney I, Piquet J, Murdaca J, Sibille B, Grimaldi PA, Neels JG, Rousseau AS. PeroxisomeProliferator Activated Receptor Beta (PPARβ) activity increases the immune response and shortens the early phases of skeletal muscle regeneration. Biochimie. 2017 May;136:33-41. doi: 10.1016/j.biochi.2016.12.001. Epub 2016 Dec 7. PMID: 27939528.
PeroxisomeProliferator Activated Receptor Beta (PPARβ) activity increases the immune response and shortens the early phases of skeletal muscle regeneration
Activation of the PPARβ pathway through the agonist GW0742 accelerates early skeletal muscle regeneration by enhancing immune cell recruitment and boosting anti-inflammatory cytokines and myogenic factors shortly after injury. In mice, this led to a quicker resolution of muscle repair markers, suggesting that PPARβ activation promotes a more efficient inflammatory and regenerative response in damaged muscle tissue.
You can read the full article at https://www.sciencedirect.com/science/article/abs/pii/S0300908416302954?via%3Dihub.
Manio MC, Inoue K, Fujitani M, Matsumura S, Fushiki T. Combined pharmacological activation of AMPK and PPARĪ“ potentiates the effects of exercise in trained mice. Physiol Rep. 2016 Mar;4(5):e12625. doi: 10.14814/phy2.12625. PMID: 26997622; PMCID: PMC4823600.
Combined pharmacological activation of AMPK and PPARĪ“ potentiates the effects of exercise in trained mice
Combined activation of AMPK and PPARĪ“ through AICAR and GW0742, alongside exercise training, significantly enhances endurance and muscle performance in mice by promoting a shift from carbohydrate to fat utilization, increasing energy substrate availability, and inducing transcriptional changes in muscle and liver. This dual pharmacological approach mimics and amplifies exercise benefits, leading to greater endurance, delayed hypoglycemia, and improved energy metabolism.
You can read the full article at https://pmc.ncbi.nlm.nih.gov/articles/PMC4823600/.
Narkar VA, Downes M, Yu RT, Embler E, Wang YX, Banayo E, Mihaylova MM, Nelson MC, Zou Y, Juguilon H, Kang H, Shaw RJ, Evans RM. AMPK and PPARdeltaagonists are exercise mimetics. Cell. 2008 Aug 8;134(3):405-15. doi: 10.1016/j.cell.2008.06.051. Epub 2008 Jul 31. PMID: 18674809; PMCID: PMC2706130.
AMPK and PPARdeltaagonists are exercise mimetics
This study shows that activating the AMPK-PPARĪ“ pathway with orally active drugs like AICAR and GW0742 can mimic or enhance the effects of endurance exercise, increasing oxidative muscle fibers and running performance in miceāeven without physical trainingāhighlighting their potential as therapeutic agents for improving metabolic health.
You can read the full article at https://pmc.ncbi.nlm.nih.gov/articles/PMC2706130/.
GaudelC , Schwartz C , Giordano C ,AbumradNA , Grimaldi P. 2008. Pharmacological activation of PPARβ promotes rapid and calcineurin-dependent fiber remodeling and angiogenesis in mouse skeletal muscle. Am J Physiol Endocrinol Metab295:E297āE304.
Pharmacological activation of PPARβ promotes rapid and calcineurin-dependent fiber remodeling and angiogenesis in mouse skeletal muscle
PPARβ agonists rapidly enhance muscle morphology in adult mice by increasing oxidative myofiber and capillary numbers within two days, driven by transient upregulation of myogenic and angiogenic markers through a calcineurin-dependent pathway, highlighting their potential to boost muscle endurance and vascularization.
You can read the full article at https://pmc.ncbi.nlm.nih.gov/articles/PMC4043180/.
Giordano C , Rousseau AS , Wagner N ,GaudelC ,MurdacaJ , Jehl-PiĆ©triC , Sibille B , Grimaldi PA , Lopez P. 2009. Peroxisome proliferator-activated receptor β activation promotes myonuclear accretion in skeletal muscle of adult and aged mice. Pflugers Arch 458:901ā913.
Peroxisome proliferator-activated receptor β activation promotes myonuclear accretion in skeletal muscle of adult and aged mice
PPARβ activation promotes exercise-like muscle remodeling by increasing oxidative fibers, capillaries, and myonuclear density through a calcineurin-dependent pathway, primarily by enhancing fusion of existing muscle precursor cells rather than new cell proliferation, and can counteract age-related declines in muscle structure and function.
You can read the full article at https://pmc.ncbi.nlm.nih.gov/articles/PMC2719750/.
Wagner, N., Jehl-PiĆ©tri, C., Lopez, P., Murdaca, J., Giordano, C., Schwartz, C., Gounon, P., Hatem, S. N., Grimaldi, P., & Wagner, K. D. (2009). Peroxisome proliferator-activated receptor beta stimulation induces rapid cardiac growth and angiogenesis via direct activation of calcineurin. Cardiovascular research, 83(1), 61ā71. https://doi.org/10.1093/cvr/cvp106.
Peroxisome proliferator-activated receptor beta stimulation induces rapid cardiac growth and angiogenesis via direct activation of calcineurin
Pharmacological activation of PPARβ using GW0742 or GW501516 enhances cardiac vascularization and increases heart size through cardiomyocyte enlargement without causing pathological hypertrophy, and these effects are mediated via activation of the calcineurin A (CnA) signaling pathway, highlighting PPARβ as a potential therapeutic target for promoting healthy cardiac growth and vascularization.
You can read the abstract of the article at https://academic.oup.com/cardiovascres/article-abstract/83/1/61/312611?redirectedFrom=fulltext&login=false.
Allen, D. M., Chen, L. E., Seaber, A. V., & Urbaniak, J. R. (1995). Pathophysiology and related studies of the no reflow phenomenon in skeletal muscle. Clinical orthopaedics and related research, (314), 122ā133.
Pathophysiology and related studies of the no reflow phenomenon in skeletal muscle
Despite advances in microvascular surgery, some tissues fail to reperfuse due to a phenomenon called “no reflow,” primarily caused by intracellular calcium overload, oxygen-free radical damage, and disrupted arachidonic acid metabolism, all of which first damage the endothelium; while several pharmacologic agents show promise in preventing no reflow, further research is needed for clinical application.
You can read the abstract of the article at https://pubmed.ncbi.nlm.nih.gov/7634624/.
Roy, R. R., Monke, S. R., Allen, D. L., & Edgerton, V. R. (1999). Modulation of myonuclear number in functionally overloaded and exercised rat plantaris fibers. Journal of applied physiology (Bethesda, Md. : 1985), 87(2), 634ā642. https://doi.org/10.1152/jappl.1999.87.2.634.
Modulation of myonuclear number in functionally overloaded and exercised rat plantaris fibers
A 10-week study on functional overload (with or without endurance training) in rat plantaris muscle showed significant increases in fiber cross-sectional area and myonuclear number across all fiber types, while maintaining consistent myonuclear domain size, suggesting that muscle hypertrophy involves proportional myonuclear addition to support increased fiber volume and preserve efficient genetic regulation.
You can read the full article at https://journals.physiology.org/doi/full/10.1152/jappl.1999.87.2.634?rfr_dat=cr_pub++0pubmed&url_ver=Z39.88-2003&rfr_id=ori%3Arid%3Acrossref.org.
Li, P., Akimoto, T., Zhang, M., Williams, R. S., & Yan, Z. (2006). Resident stem cells are not required for exercise-induced fiber-type switching and angiogenesis but are necessary for activity-dependent muscle growth. American journal of physiology. Cell physiology, 290(6), C1461āC1468. https://doi.org/10.1152/ajpcell.00532.2005.
Resident stem cells are not required for exercise-induced fiber-type switching and angiogenesis but are necessary for activity-dependent muscle growth
Endurance exercise induces significant remodeling in skeletal muscle, including fiber-type switching and angiogenesis, with a marked increase in cell proliferation. While this proliferation is not necessary for fiber-type transitions or new blood vessel formation, it is essential for muscle growth, as blocking stem cell activity halts muscle mass increases despite continued angiogenesis and fiber adaptation.
You can read the full article at https://journals.physiology.org/doi/full/10.1152/ajpcell.00532.2005?rfr_dat=cr_pub++0pubmed&url_ver=Z39.88-2003&rfr_id=ori%3Arid%3Acrossref.org.
Xiao HB, Sui GG, Lu XY. Phillyrin lowers body weight in obese mice via the modulation of PPAR/-ANGPTL 4 pathway. Obes Res Clin Pract. 2018 Jan-Feb;12(Suppl 2):71-79. doi: 10.1016/j.orcp.2017.02.002. Epub 2017 Mar 17. PMID: 28320596.
Phillyrin lowers body weight in obese mice via the modulation of PPAR/-ANGPTL 4 pathway
Phillyrin, a natural glucoside, significantly reduced body weight, liver fat, and inflammation markers in obese mice, showing effects similar to the PPARβ/Ī“ agonist GW0742. These benefits were associated with increased expression of PPARβ/Ī“, ANGPTL4, and activated AMPK, suggesting that phillyrin may help combat obesity by targeting the PPARβ/Ī“āANGPTL4 pathway.
You can read the abstract of the article at https://www.sciencedirect.com/science/article/abs/pii/S1871403X17300157?via%3Dihub.
Harrington WW, S Britt C, G Wilson J, et al. The Effect of PPARalpha, PPARdelta, PPARgamma, and PPARpan Agonists on Body Weight, Body Mass, and Serum Lipid Profiles in Diet-Induced Obese AKR/J Mice. PPAR Res. 2007;2007:97125. doi:10.1155/2007/97125.
The Effect of PPARalpha, PPARdelta, PPARgamma, and PPARpan Agonists on Body Weight, Body Mass, and Serum Lipid Profiles in Diet-Induced Obese AKR/J Mice
Activation of PPARα and PPARΓ subtypes promotes fat loss, while PPARγ activation alone leads to weight gain due to increased food intake. However, combining PPARα and PPARΓ agonists or using PPARpan agonists (targeting all three PPARs) can reduce body weight and fat mass effectively, with PPARpan compounds offering antidiabetic benefits without the weight gain typically associated with PPARγ agonists.
You can read the full article at https://pmc.ncbi.nlm.nih.gov/articles/PMC1940322/.
Toral M, Gómez-GuzmĆ”n M, JimĆ©nez R, Romero M, Zarzuelo MJ, Utrilla MP, Hermenegildo C, Cogolludo Ć, PĆ©rez-VizcaĆno F, GĆ”lvez J, Duarte J. Chronic peroxisomeproliferator-activated receptorβ/Ī“ agonist GW0742 prevents hypertension, vascular inflammatory and oxidative status, and endothelial dysfunction in diet-induced obesity. J Hypertens. 2015 Sep;33(9):1831-44. doi: 10.1097/HJH.0000000000000634. PMID: 26147382.
Chronic peroxisomeproliferator-activated receptorβ/Γ agonist GW0742 prevents hypertension, vascular inflammatory and oxidative status, and endothelial dysfunction in diet-induced obesity
Chronic activation of PPARβ/Ī“ with the agonist GW0742 in high-fat dietāfed obese mice prevents weight gain, organ hypertrophy, and fat accumulation, while improving insulin sensitivity, lipid profile, and endothelial function. GW0742 restores nitric oxideādependent vasodilation and reduces vascular oxidative stress and inflammation through enhanced Akt and eNOS signaling and decreased expression of pro-inflammatory markers. These protective effects are blocked by the PPARβ/Ī“ antagonist GSK0660, highlighting PPARβ/Ī“ās therapeutic potential in preventing obesity-related cardiovascular dysfunction.
You can read the abstract of the article at https://journals.lww.com/jhypertension/abstract/2015/09000/chronic_peroxisome_proliferator_activated.15.aspx.
Harrington WW, S Britt C, G Wilson J, et al. The Effect of PPARalpha, PPARdelta, PPARgamma, and PPARpan Agonists on Body Weight, Body Mass, and Serum Lipid Profiles in Diet-Induced Obese AKR/J Mice. PPAR Res. 2007;2007:97125. doi:10.1155/2007/97125.
The Effect of PPARalpha, PPARdelta, PPARgamma, and PPARpan Agonists on Body Weight, Body Mass, and Serum Lipid Profiles in Diet-Induced Obese AKR/J Mice
Activation of PPARα and PPARΓ subtypes promotes fat loss and enhances fatty acid oxidation in obese mice, while PPARγ agonists increase fat mass due to higher food intake. Combined activation of PPARα and Γ leads to synergistic weight and fat mass reduction. PPARpan agonists, which target all three subtypes, show strong antidiabetic effects without the weight gain typical of PPARγ agonists alone. Notably, the efficacy of PPARpan agonists depends on strong PPARΓ activation, highlighting the critical role of PPARα/Γ in driving weight loss while preserving insulin sensitivity.
You can read the full article at https://pmc.ncbi.nlm.nih.gov/articles/PMC1940322/.
Harrington, W. W., S Britt, C., G Wilson, J., O Milliken, N., G Binz, J., C Lobe, D., R Oliver, W., C Lewis, M., & M Ignar, D. (2007). The Effect of PPARalpha, PPARdelta, PPARgamma, and PPARpan Agonists on Body Weight, Body Mass, and Serum Lipid Profiles in Diet-Induced Obese AKR/J Mice. PPAR research, 2007, 97125. https://doi.org/10.1155/2007/97125.
The Effect of PPARalpha, PPARdelta, PPARgamma, and PPARpan Agonists on Body Weight, Body Mass, and Serum Lipid Profiles in Diet-Induced Obese AKR/J Mice
Activation of PPARα and PPARΓ subtypes promotes fat loss and enhances fatty acid oxidation in obese mice, while PPARγ agonists increase fat mass due to higher food intake. Combined activation of PPARα and Γ leads to synergistic weight and fat mass reduction. PPARpan agonists, which target all three subtypes, show strong antidiabetic effects without the weight gain typical of PPARγ agonists alone. Notably, the efficacy of PPARpan agonists depends on strong PPARΓ activation, highlighting the critical role of PPARα/Γ in driving weight loss while preserving insulin sensitivity.
You can read the full article at https://pmc.ncbi.nlm.nih.gov/articles/PMC1940322/.
Rachid, T. L., Silva-Veiga, F. M., Graus-Nunes, F., Bringhenti, I., Mandarim-de-Lacerda, C. A., & Souza-Mello, V. (2018). Differential actions of PPAR-α and PPAR-β/Γ on beige adipocyte formation: A study in the subcutaneous white adipose tissue of obese male mice. PloS one, 13(1), e0191365. https://doi.org/10.1371/journal.pone.0191365.
Differential actions of PPAR-α and PPAR-β/Γ on beige adipocyte formation: A study in the subcutaneous white adipose tissue of obese male mice
This study shows that while both PPAR-α and PPAR-β/Γ agonists improve metabolic health in obese mice by reducing adipocyte hypertrophy and improving glucose tolerance, only PPAR-α activation induces browning of subcutaneous white adipose tissue, marked by increased UCP1 expression and thermogenesis. In contrast, PPAR-β/Γ improves metabolism mainly through enhanced β-oxidation without promoting beige cell formation, highlighting PPAR-α as a promising target for obesity treatment via thermogenic reprogramming of fat.
You can read the full article at https://pmc.ncbi.nlm.nih.gov/articles/PMC5774787/.
Cheng KC, Chang WT, Li Y, Cheng YZ, Cheng JT, Chen ZC. GW0742 activates peroxisomeproliferator-activated receptor Ī“ to reduce free radicals and alleviate cardiac hypertrophy induced by hyperglycemia in cultured H9c2 cells. J Cell Biochem. 2018 Nov;119(11):9532-9542. doi: 10.1002/jcb.27270. Epub 2018 Aug 20. PMID: 30129179.
GW0742 activates peroxisomeproliferator-activated receptor Ī“ to reduce free radicals and alleviate cardiac hypertrophy induced by hyperglycemia in cultured H9c2 cells
Activation of PPARΓ by the agonist GW0742 inhibits cardiac hypertrophy in high-glucose-treated H9c2 cells by reducing cellular calcium levels, oxidative stress, and hypertrophic signaling pathways such as calcineurin and NFAT. This leads to lower expression of hypertrophic markers like BNP and β-MHC. These effects are reversed by the PPARΓ antagonist GSK0660, confirming the role of PPARΓ activation. Additionally, GW0742 boosts antioxidant defenses through upregulation of thioredoxin, highlighting PPARΓ as a potential therapeutic target for treating cardiac hypertrophy.
You can read the abstract of the article at https://onlinelibrary.wiley.com/doi/10.1002/jcb.27270.
Galatou E, Kelly T, Lazou A. The PPARβ/Γ agonist GW0742 modulates signaling pathways associated with cardiac myocyte growth via a non-genomic redox mechanism. Mol Cell Biochem. 2014 Oct;395(1-2):145-54. doi: 10.1007/s11010-014-2120-5. Epub 2014 Jun 18. PMID: 24939361.
The PPARβ/Γ agonist GW0742 modulates signaling pathways associated with cardiac myocyte growth via a non-genomic redox mechanism
PPARβ/Γ activation by the selective agonist GW0742 attenuates cardiac hypertrophy by inhibiting key growth signaling pathways, including ERK1/2 and PI3K/Akt, in cardiac myocytes. This effect occurs through a non-genomic mechanism involving the prevention of reactive oxygen species (ROS) generation and the preservation of PTEN phosphatase activity, rather than direct PPARβ/Γ activation, as it is not reversed by a PPARβ/Γ antagonist. These findings suggest GW0742 modulates phosphatase activity to suppress hypertrophic signaling, offering a potential therapeutic strategy for controlling cardiac hypertrophy.
You can read the abstract of the article at https://link.springer.com/article/10.1007/s11010-014-2120-5.
Kojonazarov B, Luitel H, Sydykov A, et al. The peroxisome proliferator-activated receptor β/Γ agonist GW0742 has direct protective effects on right heart hypertrophy [published correction appears in Pulm Circ. 2014 Jun;4(2):352]. Pulm Circ. 2013;3(4):926-935. doi:10.1086/674755.
The peroxisome proliferator-activated receptor β/Γ agonist GW0742 has direct protective effects on right heart hypertrophy
The PPARβ/Ī“ agonist GW0742 provides direct protective effects on the right heart in a mouse model of pulmonary hypertension induced by pulmonary artery banding (PAB), reducing hypertrophy, failure, and fibrosis without affecting vascular remodeling. Transcriptomic analysis revealed significant gene expression changes, notably increased Angptl4 expression, suggesting a molecular mechanism for GW0742’s benefits. These findings highlight PPARβ/Ī“ as a promising therapeutic target for treating right heart complications in pulmonary hypertension, potentially complementing existing vasodilator therapies.
You can read the full article at https://pmc.ncbi.nlm.nih.gov/articles/PMC4070821/.
Harrington LS, Moreno L, Reed A, et al. The PPARbeta/delta agonist GW0742 relaxes pulmonary vessels and limits right heart hypertrophy in rats with hypoxia-induced pulmonary hypertension. PLoS One. 2010;5(3):e9526. Published 2010 Mar 4. doi:10.1371/journal.pone.0009526.
The PPARbeta/delta agonist GW0742 relaxes pulmonary vessels and limits right heart hypertrophy in rats with hypoxia-induced pulmonary hypertension
The PPARβ/Γ agonist GW0742 induces vasorelaxation in both systemic and pulmonary vessels and significantly reduces right heart hypertrophy and systolic pressure in a rat model of hypoxia-induced pulmonary hypertension, despite not affecting vascular remodeling. Interestingly, its vasodilatory effects are independent of PPARβ/Γ or prostacyclin receptors and are instead linked to RhoA inhibition. These findings highlight GW0742 as a promising therapeutic candidate for pulmonary hypertension, supporting the potential for future clinical trials.
You can read the full article at https://pmc.ncbi.nlm.nih.gov/articles/PMC2831997/.
Kuo, S. C., Ku, P. M., Chen, L. J., Niu, H. S., & Cheng, J. T. (2013). Activation of receptors Ī“ (PPARĪ“) by agonist (GW0742) may enhance lipid metabolism in heart both in vivo and in vitro. Hormone and metabolic research = Hormon- und Stoffwechselforschung = Hormones et metabolisme, 45(12), 880ā886. https://doi.org/10.1055/s-0033-1348317.
Activation of receptors Ī“ (PPARĪ“) by agonist (GW0742) may enhance lipid metabolism in heart both in vivo and in vitro
This study demonstrates that direct activation of PPARĪ“ by the agonist GW0742 enhances lipid metabolism in the heart by upregulating fatty acid oxidation and citric acid cycle-related genes in both Wistar rats and cultured cardiomyocytes. These effects were blocked by a PPARĪ“ antagonist or siRNA, confirming the specificity of the pathway. The findings establish PPARĪ“ as a key regulator of cardiac lipid metabolism.
You can read the abstract of the article at https://www.thieme-connect.com/products/ejournals/abstract/10.1055/s-0033-1348317.
Romero M, Toral M, Robles-Vera I, SĆ”nchez M, JimĆ©nez R, O’Valle F, Rodriguez-Nogales A, PĆ©rez-Vizcaino F, GĆ”lvez J, Duarte J. Activation of PeroxisomeProliferator Activator Receptor β/Ī“ Improves Endothelial Dysfunction and Protects Kidney in Murine Lupus. Hypertension. 2017 Apr;69(4):641-650. doi: 10.1161/HYPERTENSIONAHA.116.08655. Epub 2017 Feb 27. PMID: 28242713.
Activation of PeroxisomeProliferator Activator Receptor β/Γ Improves Endothelial Dysfunction and Protects Kidney in Murine Lupus. Hypertension
Activation of PPARβ/Γ with the agonist GW0742 significantly reduced disease activity and cardiovascular complications in a female mouse model of lupus, including lowering blood pressure, renal and cardiac hypertrophy, albuminuria, vascular oxidative stress, and endothelial dysfunction. GW0742 also decreased inflammatory cytokines, immune cell activation, and autoantibody levels, while increasing hepatic opsonins and normalizing spleen lymphocyte populations. These beneficial effects were blocked by a PPARβ/Γ antagonist, highlighting PPARβ/Γ as a promising therapeutic target for systemic lupus erythematosus and its associated vascular damage.
You can read the full article at https://www.ahajournals.org/doi/10.1161/HYPERTENSIONAHA.116.08655?url_ver=Z39.88-2003&rfr_id=ori:rid:crossref.org&rfr_dat=cr_pub%20%200pubmed.
Yue TL, Nerurkar SS, Bao W, Jucker BM, Sarov-Blat L, Steplewski K, Ohlstein EH, Willette RN. In vivo activation of peroxisomeproliferator-activated receptor-delta protects the heart from ischemia/reperfusion injury in Zucker fatty rats. J Pharmacol Exp Ther. 2008 May;325(2):466-74. doi: 10.1124/jpet.107.135327. Epub 2008 Feb 20. PMID: 18287212.
In vivo activation of peroxisomeproliferator-activated receptor-delta protects the heart from ischemia/reperfusion injury in Zucker fatty rats
Pharmacological activation of PPAR-delta with the selective agonist GW0742 significantly protected the hearts of obese Zucker fatty rats from ischemia/reperfusion injury by reducing infarct size, cardiac troponin-I levels, apoptosis, and inflammation. GW0742 improved cardiac function, enhanced fatty acid oxidation, lowered circulating free fatty acids, and activated prosurvival Akt signaling while attenuating mitochondrial damage and inflammatory cytokine expression. These findings suggest that PPAR-delta activation confers cardioprotection through combined metabolic, anti-inflammatory, and anti-apoptotic mechanisms.
You can read the abstract of the article at https://jpet.aspetjournals.org/article/S0022-3565(24)34408-8/abstract.
Schnegg, C. I., Greene-Schloesser, D., Kooshki, M., Payne, V. S., Hsu, F. C., & Robbins, M. E. (2013). The PPARĪ“ agonist GW0742 inhibits neuroinflammation, but does not restore neurogenesis or prevent early delayed hippocampal-dependent cognitive impairment after whole-brain irradiation. Free radical biology & medicine, 61, 1ā9. https://doi.org/10.1016/j.freeradbiomed.2013.03.002.
The PPARĪ“ agonist GW0742 inhibits neuroinflammation, but does not restore neurogenesis or prevent early delayed hippocampal-dependent cognitive impairment after whole-brain irradiation
This study found that the PPARĪ“ agonist GW0742 reduced early neuroinflammatory responses and microglial activation following whole-brain irradiation (WBI) in wild-type mice but failed to prevent long-term reductions in hippocampal neurogenesis or cognitive impairments. In PPARĪ“ knockout mice, some radiation responses were altered, suggesting a complex role for PPARĪ“ in radiation-induced brain injury. While GW0742 offers partial protection against acute inflammation, it does not mitigate the lasting cognitive and neurogenic deficits caused by WBI, highlighting the complex and not fully understood relationship between neuroinflammation, neurogenesis, and cognition.
You can read the full article at https://pmc.ncbi.nlm.nih.gov/articles/PMC3884086/.
An YQ, Zhang CT, Du Y, Zhang M, Tang SS, Hu M, Long Y, Sun HB, Hong H. PPARΓ agonist GW0742 ameliorates Aβ1-42-induced hippocampal neurotoxicity in mice. Metab Brain Dis. 2016 Jun;31(3):663-71. doi: 10.1007/s11011-016-9800-7. Epub 2016 Feb 11. PMID: 26864581.
PPARΓ agonist GW0742 ameliorates Aβ1-42-induced hippocampal neurotoxicity in mice
This study shows that activating PPARĪ“ with the agonist GW0742 can protect against amyloid-β (Aβ1-42)-induced neurotoxicity in the hippocampus of mice. GW0742 reversed memory impairments, reduced neuroinflammation and apoptosis, and restored PPARĪ“ expression levels in the hippocampus following Aβ exposure. These findings suggest that PPARĪ“ activation may be a promising therapeutic approach for mitigating cognitive deficits and neuronal damage associated with Alzheimerās disease.
You can read the abstract of the article at https://link.springer.com/article/10.1007/s11011-016-9800-7.
Benedetti E, Di Loreto S, D’Angelo B, Cristiano L, d’Angelo M, Antonosante A, Fidoamore A, Golini R, Cinque B, Cifone MG, Ippoliti R, Giordano A, Cimini A. The PPARβ/Ī“ Agonist GW0742 Induces Early Neuronal Maturation of Cortical Post-Mitotic Neurons: Role of PPARβ/Ī“ in Neuronal Maturation. J Cell Physiol. 2016 Mar;231(3):597-606. doi: 10.1002/jcp.25103. PMID: 26206209.
The PPARβ/Γ Agonist GW0742 Induces Early Neuronal Maturation of Cortical Post-Mitotic Neurons: Role of PPARβ/Γ in Neuronal Maturation
This study demonstrates that PPARβ/Ī“ plays a key role in neuronal maturation, with its activation by the agonist GW0742 accelerating neuronal differentiation in cortical neuron cultures. The effects were blocked by the PPARβ/Ī“ antagonist GSK0660, highlighting the receptorās specificity. The findings suggest that PPARβ/Ī“ promotes neuronal development, likely through activation of the BDNF signaling pathway, and may have implications for understanding learning, memory, and neurodegenerative diseases.
You can read the abstract of the article at https://onlinelibrary.wiley.com/doi/10.1002/jcp.25103.
Konttinen H, Gureviciene I, Oksanen M, Grubman A, Loppi S, Huuskonen MT, Korhonen P, Lampinen R, Keuters M, Belaya I, Tanila H, Kanninen KM, Goldsteins G, Landreth G, Koistinaho J, Malm T. PPARβ/Ī“-agonist GW0742 ameliorates dysfunction in fatty acid oxidation in PSEN1ĪE9 astrocytes. Glia. 2019 Jan;67(1):146-159. doi: 10.1002/glia.23534. Epub 2018 Nov 19. PMID: 30453390; PMCID: PMC7526864.
PPARβ/Ī“-agonist GW0742 ameliorates dysfunction in fatty acid oxidation in PSEN1ĪE9 astrocytes
This study shows that fatty acid oxidation is impaired in astrocytes derived from Alzheimer’s disease (AD) patients, contributing to neurodegeneration and memory deficits. Treatment with the PPARβ/Ī“ agonist GW0742 restored fatty acid metabolism by upregulating CPT1a expression, enhanced neurogenesis and neuronal differentiation, and improved cognitive performance in an AD mouse model. Although GW0742 did not reduce amyloid beta levels, it prevented Aβ-induced synaptic dysfunction, suggesting that targeting astrocytic energy metabolism via PPARβ/Ī“ activation could be a promising therapeutic strategy for AD.
You can read the full article at https://pmc.ncbi.nlm.nih.gov/articles/PMC7526864/.
Abdel-Rahman EA, Bhattacharya S, Buabeid M, Majrashi M, Bloemer J, Tao YX, Dhanasekaran M, Escobar M, Amin R, Suppiramaniam V. PPAR-Ī“ Activation Ameliorates Diabetes-Induced Cognitive Dysfunction by Modulating Integrin-linked Kinase and AMPA Receptor Function. J Am CollNutr. 2019 Nov-Dec;38(8):693-702. doi: 10.1080/07315724.2019.1598307. Epub 2019 Apr 22. PMID: 31008686.
PPAR-Ī“ Activation Ameliorates Diabetes-Induced Cognitive Dysfunction by Modulating Integrin-linked Kinase and AMPA Receptor Function
This study demonstrates that activation of PPAR-Ī“ with the selective agonist GW0742 improves hippocampal synaptic plasticity and spatial memory in a mouse model of type 2 diabetes (db/db mice). Oral GW0742 treatment restored long-term potentiation and AMPA receptor-mediated synaptic transmission in the hippocampus, effectively preventing cognitive deficits associated with diabetes. These findings suggest that PPAR-Ī“ activation may be a promising therapeutic strategy for mitigating T2DM-induced cognitive impairments.
You can read the abstract of the article at https://www.tandfonline.com/doi/10.1080/07315724.2019.1598307?url_ver=Z39.88-2003&rfr_id=ori:rid:crossref.org&rfr_dat=cr_pub%20%200pubmed.
Kuang G, He Q, Zhang Y, Zhuang R, Xiang A, Jiang Q, Luo Y, Yang J. Modulation of Preactivation of PPAR-β on Memory and Learning Dysfunction and Inflammatory Response in the Hippocampus in Rats Exposed to Global Cerebral Ischemia/Reperfusion. PPAR Res. 2012;2012:209794. doi: 10.1155/2012/209794. Epub 2012 Sep 27. PMID: 23056034; PMCID: PMC3465902.
Modulation of Preactivation of PPAR-β on Memory and Learning Dysfunction and Inflammatory Response in the Hippocampus in Rats Exposed to Global Cerebral Ischemia/Reperfusion
This study found that the PPAR-β agonist GW0742 offers significant neuroprotection against global cerebral ischemia-reperfusion injury (GCIRI) in rats. Pretreatment with GW0742 improved memory and hippocampal neuronal morphology, reduced proinflammatory cytokines (IL-1β, IL-6, TNF-α), and increased the anti-inflammatory cytokine IL-10. It also suppressed NF-κB p65 expression and nuclear translocation in hippocampal neurons. These findings suggest GW0742 protects the brain through PPAR-β activation and anti-inflammatory mechanisms, particularly by inhibiting NF-κB signaling.
You can read the full article at https://pmc.ncbi.nlm.nih.gov/articles/PMC3465902/.
Smith SA, Monteith GR, Robinson JA, Venkata NG, May FJ, Roberts-Thomson SJ. Effect of the peroxisomeproliferator-activated receptor beta activator GW0742 in rat cultured cerebellar granule neurons. J Neurosci Res. 2004 Jul 15;77(2):240-9. doi: 10.1002/jnr.20153. PMID: 15211590.
Effect of the peroxisomeproliferator-activated receptor beta activator GW0742 in rat cultured cerebellar granule neurons
This study explored the role of PPARβ in neuronal cell death using the ligand GW0742 in cultured rat cerebellar granule neurons. PPARβ activation by GW0742 was not acutely toxic but showed delayed apoptotic effects after 48 hours. Interestingly, GW0742 also demonstrated neuroprotective properties by significantly reducing cell death under stress conditions (low-KCl exposure). These findings highlight the distinct functions of PPAR isoforms in neurons and suggest that PPARβ may be a promising therapeutic target for neurodegenerative diseases.
You can read the abstract of the article at https://onlinelibrary.wiley.com/doi/10.1002/jnr.20153.
Benetti, E., Mastrocola, R., Rogazzo, M., Chiazza, F., Aragno, M., Fantozzi, R., Collino, M., & Minetto, M. A. (2013). High sugar intake and development of skeletal muscle insulin resistance and inflammation in mice: a protective role for PPAR- Ī“ agonism. Mediators of inflammation, 2013, 509502. https://doi.org/10.1155/2013/509502.
High sugar intake and development of skeletal muscle insulin resistance and inflammation in mice
This study shows that chronic activation of PPAR-Ī“ by the selective agonist GW0742 improves glucose and lipid metabolism in skeletal muscle of mice exposed to a high-fructose corn syrup (HFCS) diet. HFCS induced insulin resistance, inflammation, and reduced glucose transporter expression in the gastrocnemius muscle, while GW0742 treatment reversed these effects by enhancing insulin and AMPK signaling, reducing inflammation, and increasing expression of metabolic regulators like GLUT-4, GLUT-5, and fibroblast growth factor-21. These findings suggest that PPAR-Ī“ activation protects skeletal muscle from HFCS-induced metabolic dysfunction.
You can read the full article at https://pmc.ncbi.nlm.nih.gov/articles/PMC3703883/#:~:text=In%20conclusion%2C%20we%20have%20shown,by%20selective%20PPAR%2D%CE%B4%20activation.
Quintela, A. M., JimĆ©nez, R., Gómez-GuzmĆ”n, M., Zarzuelo, M. J., Galindo, P., SĆ”nchez, M., Vargas, F., Cogolludo, A., Tamargo, J., PĆ©rez-VizcaĆno, F., & Duarte, J. (2012). Activation of peroxisome proliferator-activated receptor-β/-Ī“ (PPARβ/Ī“) prevents endothelial dysfunction in type 1 diabetic rats. Free radical biology & medicine, 53(4), 730ā741. https://doi.org/10.1016/j.freeradbiomed.2012.05.045.
Activation of peroxisome proliferator-activated receptor-β/-Γ (PPARβ/Γ) prevents endothelial dysfunction in type 1 diabetic rats
This study shows that activation of PPARβ/Γ by the agonist GW0742 restores endothelial function in type 1 diabetic rats without affecting blood glucose or blood pressure. GW0742 improved acetylcholine-induced vasodilation by increasing eNOS phosphorylation and reducing oxidative stress through downregulation of NADPH oxidase components and prepro endothelin-1 expression. These protective effects were blocked by a PPARβ/Γ antagonist, indicating that PPARβ/Γ activation enhances nitric oxide bioavailability and reduces vascular oxidative damage, thereby improving endothelial health in diabetes.
You can read the full article at https://www.sciencedirect.com/science/article/abs/pii/S0891584912003024?via%3Dihub.
Tian XY, Wong WT, Wang N, et al. PPARĪ“ activation protects endothelial function in diabetic mice. Diabetes. 2012;61(12):3285-3293. doi:10.2337/db12-0117.
PPARĪ“ activation protects endothelial function in diabetic mice
This study demonstrates that activation of PPARĪ“ with the agonist GW1516 improves endothelial function in diabetic and obese mice by enhancing nitric oxide (NO) production via the PI3K/Akt/eNOS signaling pathway. Treatment with GW1516 restored endothelium-dependent relaxation and flow-mediated vasodilation in diabetic models, with effects dependent on PPARĪ“ activation. These findings highlight the therapeutic potential of PPARĪ“ agonists in improving vascular health and treating diabetic vasculopathy.
You can read the full article at https://pmc.ncbi.nlm.nih.gov/articles/PMC3501853/.
Das, N. R., Gangwal, R. P., Damre, M. V., Sangamwar, A. T., & Sharma, S. S. (2014). A PPAR-β/Ī“ agonist is neuroprotective and decreases cognitive impairment in a rodent model of Parkinson’s disease. Current neurovascular research, 11(2), 114ā124. https://doi.org/10.2174/1567202611666140318114037.
A PPAR-β/Ī“ agonist is neuroprotective and decreases cognitive impairment in a rodent model of Parkinson’s disease
This study shows that the PPAR-β/Ī“ agonist GW0742 significantly improves cognitive impairments in a rat model of Parkinsonās disease (PD) induced by MPTP, as demonstrated by enhanced memory performance and reduced oxidative damage and neuronal loss. GW0742 also restored tyrosine hydroxylase levels, indicating neuroprotection, and pharmacokinetic modeling confirmed its ability to cross the blood-brain barrier. These findings suggest that PPAR-β/Ī“ activation may be a promising therapeutic strategy for PD-related cognitive dysfunction.
You can read the abstract of the article at https://pubmed.ncbi.nlm.nih.gov/24635117/.
Malm, T., Mariani, M., Donovan, L. J., Neilson, L., & Landreth, G. E. (2015). Activation of the nuclear receptor PPARĪ“ is neuroprotective in a transgenic mouse model of Alzheimer’s disease through inhibition of inflammation. Journal of neuroinflammation, 12, 7. https://doi.org/10.1186/s12974-014-0229-9.
Activation of the nuclear receptor PPARĪ“ is neuroprotective in a transgenic mouse model of Alzheimer’s disease through inhibition of inflammation
This study demonstrates that the PPARĪ“ agonist GW0742 reduces beta-amyloid plaque burden, suppresses neuroinflammation, and preserves neuronal survival in a mouse model of Alzheimerās disease (5XFAD). GW0742 treatment decreased proinflammatory microglial activation while enhancing their presence around plaques, suggesting a shift to a more beneficial, phagocytic phenotype. Although GW0742 did not protect neurons from direct glutamate toxicity, it prevented inflammation-induced neuronal death in co-culture models. These findings highlight the therapeutic potential of PPARĪ“ agonists in mitigating neurodegeneration in AD through anti-inflammatory mechanisms.
You can read the full article at https://jneuroinflammation.biomedcentral.com/articles/10.1186/s12974-014-0229-9.
Romero M, JimĆ©nez R, Toral M, León-Gómez E, Gómez-GĆŗzman M, SĆ”nchez M, Zarzuelo MJ, RodrĆguez-Gómez I, Rath G, Tamargo J, PĆ©rez-VizcaĆno F, Dessy C, Duarte J. Vascular and Central Activation of PeroxisomeProliferator-Activated Receptor-β Attenuates Angiotensin II-Induced Hypertension: Role of RGS-5. J Pharmacol Exp Ther. 2016 Jul;358(1):151-63. doi: 10.1124/jpet.116.233106. Epub 2016 Apr 27. PMID: 27189971.
Vascular and Central Activation of PeroxisomeProliferator-Activated Receptor-β Attenuates Angiotensin II-Induced Hypertension: Role of RGS-5
This study shows that activation of PPARβ by the agonist GW0742 prevents high blood pressure in a mouse model of angiotensin II-induced hypertension by restoring vascular and neural function. GW0742 reduced vascular remodeling, sympathetic overactivity, and vasoconstrictor responsiveness, while suppressing oxidative stress and increasing expression of RGS5, a negative regulator of angiotensin II signaling. These protective effects were blocked by a PPARβ antagonist or RGS5 knockdown, highlighting that GW0742 lowers blood pressure through PPARβ-dependent upregulation of RGS5 in both blood vessels and brain.
You can read the abstract of the article at https://linkinghub.elsevier.com/retrieve/pii/S0022-3565(24)19341-X.
MarĆa JosĆ© Zarzuelo, Manuel Gómez-GuzmĆ”n, Rosario JimĆ©nez, Ana MarĆa Quintela, Miguel Romero, Manuel SĆ”nchez, Antonio Zarzuelo, Juan Tamargo, Francisco PĆ©rez-VizcaĆno, Juan Duarte, Effects of peroxisome proliferator-activated receptor-β activation in endothelin-dependent hypertension, Cardiovascular Research, Volume 99, Issue 4, 1 September 2013, Pages 622ā631, https://doi.org/10.1093/cvr/cvt152.
Effects of peroxisome proliferator-activated receptor-β activation in endothelin-dependent hypertension
This study shows that the PPAR-β agonist GW0742 prevents hypertension and vascular damage in a rat model of DOCA-salt-induced, renin-independent hypertension. While GW0742 lowered blood pressure through both PPAR-β-dependent and -independent mechanisms, its vascular protective effectsāsuch as reduced oxidative stress, improved endothelial function, and decreased endothelin-1 signalingāwere PPAR-β-dependent and involved upregulation of antioxidant genes and RGS5. These benefits were dose-dependent and blocked by a PPAR-β antagonist, indicating that GW0742 improves vascular health primarily through PPAR-β activation.
You can read the abstract of the article at https://academic.oup.com/cardiovascres/article-abstract/99/4/622/1746666?redirectedFrom=fulltext&login=false.
Zarzuelo MJ, JimĆ©nez R, Galindo P, SĆ”nchez M, Nieto A, Romero M, Quintela AM, López-SepĆŗlveda R, Gómez-GuzmĆ”n M, Bailón E, RodrĆguez-Gómez I, Zarzuelo A, GĆ”lvez J, Tamargo J, PĆ©rez-VizcaĆno F, Duarte J. Antihypertensive effects of peroxisome proliferator-activated receptor-β activation in spontaneously hypertensive rats. Hypertension. 2011 Oct;58(4):733-43. doi: 10.1161/HYPERTENSIONAHA.111.174490. Epub 2011 Aug 8. PMID: 21825230.
Antihypertensive effects of peroxisome proliferator-activated receptor-β activation in spontaneously hypertensive rats
This study demonstrates that activation of PPARβ by the agonist GW0742 significantly lowers blood pressure and improves vascular health in spontaneously hypertensive rats (SHRs). GW0742 reduced arterial remodeling, oxidative stress, and inflammation while enhancing endothelial nitric oxide synthase activity and downregulating harmful pathways like NADPH oxidase and ERK1/2. These effects were specific to hypertensive rats and linked to increased expression of RGS4 and RGS5, which counteract angiotensin II signaling. The findings suggest PPARβ activation as a promising therapeutic strategy for treating genetic hypertension.
You can read the full article at https://www.ahajournals.org/doi/10.1161/HYPERTENSIONAHA.111.174490?url_ver=Z39.88-2003&rfr_id=ori:rid:crossref.org&rfr_dat=cr_pub%20%200pubmed.
Niu HS, Ku PM, Niu CS, Cheng JT, Lee KS. Development of PPAR-agonist GW0742 as antidiabetic drug: study in animals. Drug Des DevelTher. 2015 Oct 14;9:5625-32. doi: 10.2147/DDDT.S95045. PMID: 26508837; PMCID: PMC4610778.
Development of PPAR-agonist GW0742 as antidiabetic drug: study in animals
This study shows that the PPAR-Ī“ agonist GW0742 improves systemic insulin sensitivity and glucose homeostasis in diabetic rats. GW0742 reduced insulin resistance, as evidenced by lower HOMA-IR scores and improved results in hyperinsulinemic euglycemic clamp tests, effects that were blocked by a PPAR-Ī“ antagonist. It also decreased hyperglycemia in type 1 diabetic rats and favorably modulated GLUT4 and PEPCK expression in skeletal muscle and liver. These findings support PPAR-Ī“ as a promising therapeutic target for diabetes treatment.
You can read the full article at https://pmc.ncbi.nlm.nih.gov/articles/PMC4610778/#:~:text=The%20data%20showed%20that%20GW0742,antidiabetic%20drugs%20in%20the%20future.
Khazaei M, Salehi E, Rashidi B, Javanmard SH, Fallahzadeh AR. Role of peroxisomeproliferator-activated receptor β agonist on angiogenesis in hindlimb ischemic diabetic rats. J Diabetes Complications. 2012 Mar-Apr;26(2):137-40. doi: 10.1016/j.jdiacomp.2012.02.005. Epub 2012 Mar 29. PMID: 22464549.
Role of peroxisomeproliferator-activated receptor β agonist on angiogenesis in hindlimb ischemic diabetic rats
This study found that the PPARβ agonist GW0742 enhances angiogenesis in both normal and diabetic rats with hindlimb ischemia. GW0742 increased serum nitrite, VEGFR-2 levels, and the VEGF-to-VEGFR-2 ratio, while significantly restoring capillary density in skeletal muscle, especially in diabetic rats where it was reduced. These findings suggest that PPARβ activation may help prevent or treat peripheral vascular complications in diabetes by promoting angiogenesis.
You can read the abstract of the article at https://www.sciencedirect.com/science/article/abs/pii/S1056872712000074?via%3Dihub.
Abdel-Rahman EA, Bhattacharya S, Buabeid M, Majrashi M, Bloemer J, Tao YX, Dhanasekaran M, Escobar M, Amin R, Suppiramaniam V. PPAR-Ī“ Activation Ameliorates Diabetes-Induced Cognitive Dysfunction by Modulating Integrin-linked Kinase and AMPA Receptor Function. J Am CollNutr. 2019 Nov-Dec;38(8):693-702. doi: 10.1080/07315724.2019.1598307. Epub 2019 Apr 22. PMID: 31008686.
PPAR-Ī“ Activation Ameliorates Diabetes-Induced Cognitive Dysfunction by Modulating Integrin-linked Kinase and AMPA Receptor Function
This study shows that the PPAR-Ī“ agonist GW0742 improves hippocampal synaptic plasticity and spatial memory in a type 2 diabetes (T2DM) mouse model (db/db mice). Two weeks of oral GW0742 treatment enhanced long-term potentiation and prevented memory deficits, likely through restoring AMPA receptor-mediated synaptic transmission in the hippocampus. These findings suggest PPAR-Ī“ activation as a potential therapeutic strategy for T2DM-related cognitive impairment.
You can read the abstract of the article at https://www.tandfonline.com/doi/10.1080/07315724.2019.1598307?url_ver=Z39.88-2003&rfr_id=ori:rid:crossref.org&rfr_dat=cr_pub%20%200pubmed.
Perez-Diaz N, Pushkarsky I, Oweis N, Lione LA, Mackenzie LS. The Non-Genomic Effects of the PPARβγ Agonist GW0742 on Streptozotocin Treated Rat Aorta. Curr Mol Pharmacol. 2018;11(2):149-154. doi: 10.2174/1874467209666161229124356. PMID: 28034288.
The Non-Genomic Effects of the PPARβγ Agonist GW0742 on Streptozotocin Treated Rat Aorta
This study reveals that the PPARβ/Ī“ agonist GW0742 can induce vasodilation in rat aorta through mechanisms that are partly independent of PPARβ/Ī“ activation and remain effective in both normal and diabetic states. Using ROCK inhibitors, researchers found that GW0742 inhibits vascular contraction responses and that its vasodilatory effect involves different pathways in diabetic versus non-diabetic conditions. These findings highlight GW0742’s potential to modulate vascular tone via off-target mechanisms beyond PPARβ/Ī“.
You can read the abstract of the article at https://www.eurekaselect.com/article/80655.
KrƤmer DK, Al-Khalili L, Perrini S, Skogsberg J, Wretenberg P, Kannisto K, Wallberg-Henriksson H, Ehrenborg E, Zierath JR, Krook A. Direct activation of glucose transport in primary human myotubes after activation of peroxisomeproliferator-activated receptor delta. Diabetes. 2005 Apr;54(4):1157-63. doi: 10.2337/diabetes.54.4.1157. PMID: 15793256.
Direct activation of glucose transport in primary human myotubes after activation of peroxisomeproliferator-activated receptor delta
This study shows that PPARĪ“ agonists (GW501516 and GW0742) enhance glucose uptake in skeletal muscle cells independently of insulin and improve insulin sensitivity, likely through activation of AMP-activated protein kinase (AMPK) and MAPK pathways, particularly p38 MAPK. These effects occurred without changes to Akt signaling and involved reduced expression of certain metabolic regulators like SREBP-1. The findings support PPARĪ“ activation as a promising strategy to improve glucose metabolism and highlight its potential as a therapeutic target for diabetes.
You can read the full article at https://diabetesjournals.org/diabetes/article/54/4/1157/14877/Direct-Activation-of-Glucose-Transport-in-Primary.
Lee MY, Choi R, Kim HM, Cho EJ, Kim BH, Choi YS, Naowaboot J, Lee EY, Yang YC, Shin JY, Shin YG, Chung CH. Peroxisomeproliferator-activated receptor Ī“ agonist attenuates hepatic steatosis by anti-inflammatory mechanism. Exp Mol Med. 2012 Oct 31;44(10):578-85. doi: 10.3858/emm.2012.44.10.066. PMID: 22824914; PMCID: PMC3490079.
Peroxisomeproliferator-activated receptor Ī“ agonist attenuates hepatic steatosis by anti-inflammatory mechanism
This study shows that PPARĪ“ agonists (GW501516 and GW0742) enhance glucose uptake in skeletal muscle cells independently of insulin and improve insulin sensitivity, likely through activation of AMP-activated protein kinase (AMPK) and MAPK pathways, particularly p38 MAPK. These effects occurred without changes to Akt signaling and involved reduced expression of certain metabolic regulators like SREBP-1. The findings support PPARĪ“ activation as a promising strategy to improve glucose metabolism and highlight its potential as a therapeutic target for diabetes.
You can read the full article at https://pmc.ncbi.nlm.nih.gov/articles/PMC3490079/.
Matsushita Y, Ogawa D, Wada J, et al. Activation of peroxisomeproliferator-activated receptor delta inhibits streptozotocin-induced diabetic nephropathy through anti-inflammatory mechanisms in mice. Diabetes. 2011;60(3):960-968. doi:10.2337/db10-1361.
Activation of peroxisomeproliferator-activated receptor delta inhibits streptozotocin-induced diabetic nephropathy through anti-inflammatory mechanisms in mice
Activation of PPARĪ“ with the agonist GW0742 provides renoprotective effects in streptozotocin-induced diabetic mice by reducing urinary albumin excretion, inflammation, and kidney damage without affecting blood glucose levels. GW0742 suppressed inflammatory mediators like MCP-1 and OPN, likely via upregulation of the anti-inflammatory corepressor B-cell lymphoma-6. These results suggest that PPARĪ“ agonists may represent a promising therapeutic option for diabetic nephropathy.
You can read the full article at https://pmc.ncbi.nlm.nih.gov/articles/PMC3046857/.
Yuichi Matsushita, Daisuke Ogawa, Jun Wada, Noriko Yamamoto, Kenichi Shikata, Chikage Sato, Hiromi Tachibana, Noriko Toyota, Hirofumi MakinoDiabetes Mar 2011, 60 (3) 960-968; DOI: 10.2337/db10-136.
Di Paola R, Crisafulli C, Mazzon E, Esposito E, Paterniti I, Galuppo M, Genovese T, Thiemermann C, Cuzzocrea S. GW0742, a high-affinity PPAR -beta/delta agonist, inhibits acute lung injury in mice. Shock. 2010 Apr;33(4):426-35. doi: 10.1097/SHK.0b013e3181b8f2fb. PMID: 20407409.
GW0742, a high-affinity PPAR -beta/delta agonist, inhibits acute lung injury in mice
The PPAR-β/Γ agonist GW0742 significantly reduces lung inflammation and injury in mice induced by bleomycin, a model for pulmonary fibrosis. GW0742 treatment lowered inflammatory cytokine production, neutrophil infiltration, oxidative stress, NF-κB activation, and apoptotic signaling, demonstrating strong anti-inflammatory and anti-apoptotic effects. These findings support GW0742 as a potential therapeutic agent for preventing lung damage and fibrosis.
You can read the abstract of the article at https://www.medchemexpress.cn/mce_publications/21244753.html.
Ozawa C, Horiguchi M, Akita T, Oiso Y, Abe K, Motomura T, Yamashita C. Pulmonary Administration of GW0742, a High-Affinity PeroxisomeProliferator-Activated Receptor Agonist, Repairs Collapsed Alveoli in an Elastase-Induced Mouse Model of Emphysema. BiolPharm Bull. 2016;39(5):778-85. doi: 10.1248/bpb.b15-00909. PMID: 27150147.
Pulmonary Administration of GW0742, a High-Affinity PeroxisomeProliferator-Activated Receptor Agonist, Repairs Collapsed Alveoli in an Elastase-Induced Mouse Model of Emphysema
The high-affinity PPARβ/Γ agonist GW0742 significantly reverses lung tissue damage and improves respiratory function in a mouse model of emphysema. By enhancing the expression of alveolar type II cell markers and reducing alveolar wall damage, GW0742 restored lung structure and improved pulmonary mechanics. These findings suggest that PPARβ/Γ activation may offer a promising therapeutic approach for treating pulmonary emphysema.
You can read the abstract of the article at https://www.jstage.jst.go.jp/article/bpb/39/5/39_b15-00909/_article.
Bao, X. C., Fang, Y. Q., You, P., Zhang, S., & Ma, J. (2014). Protective role of peroxisome proliferator-activated receptor-β/Ī“ against pulmonary oxygen toxicity mediated through changes in NOS expression levels. Experimental lung research, 40(3), 105ā116. https://doi.org/10.3109/01902148.2013.879497.
Protective role of peroxisome proliferator-activated receptor-β/Γ against pulmonary oxygen toxicity mediated through changes in NOS expression levels
This study shows that activating PPAR-β/Ī“ with the agonist GW0742 protects against lung injury caused by hyperbaric oxygen exposure in rats by reducing inflammation, lung fluid accumulation, and nitric oxide synthase (NOS) expression. In contrast, blocking PPAR-β/Ī“ with the antagonist GSK0660 worsens lung damage. Findings from knockout mice further confirm that PPAR-β/Ī“’s protective effects are linked to its regulation of NOS pathways, highlighting its potential as a therapeutic target for pulmonary oxygen toxicity.
You can read the abstract of the article at https://www.tandfonline.com/doi/10.3109/01902148.2013.879497?url_ver=Z39.88-2003&rfr_id=ori:rid:crossref.org&rfr_dat=cr_pub%20%200pubmed.
Galuppo M, Di Paola R, Mazzon E, Esposito E, Paterniti I, Kapoor A, Thiemermann C, Cuzzocrea S. GW0742, a high affinity PPAR-β/Γ agonist reduces lung inflammation induced by bleomycin instillation in mice. Int J ImmunopatholPharmacol. 2010 Oct-Dec;23(4):1033-46. doi: 10.1177/039463201002300408. PMID: 21244753.
GW0742, a high affinity PPAR-β/Γ agonist reduces lung inflammation induced by bleomycin instillation in mice
This study demonstrates that the PPAR β/Ī“ agonist GW0742 significantly reduces lung inflammation and injury caused by bleomycin in mice. GW0742 treatment decreased cytokine production, neutrophil infiltration, oxidative stress, and markers of apoptosis, suggesting its strong anti-inflammatory and anti-apoptotic effects. These findings highlight GW0742ās potential as a therapeutic agent to prevent or mitigate pulmonary fibrosis and lung damage associated with acute inflammation.
You can read the abstract of the article at https://journals.sagepub.com/doi/10.1177/039463201002300408?url_ver=Z39.88-2003&rfr_id=ori:rid:crossref.org&rfr_dat=cr_pub%20%200pubmed.
Choudhary M, Ding JD, Qi X, Boulton ME, Yao PL, Peters JM, Malek G. PPARβ/Γ selectively regulates phenotypic features of age-related macular degeneration. Aging (Albany NY). 2016 Sep 8;8(9):1952-1978. doi: 10.18632/aging.101031. PMID: 27622388; PMCID: PMC5076447.
PPARβ/Γ selectively regulates phenotypic features of age-related macular degeneration
This study reveals that PPARβ/Γ, a nuclear receptor involved in key cellular processes, plays a dual role in age-related macular degeneration (AMD). Activation of PPARβ/Γ reduces lipid accumulation in retinal pigment epithelial cells, potentially benefiting dry AMD, while its inhibition suppresses angiogenesis, relevant to wet AMD. Genetic ablation of PPARβ/Γ worsened dry AMD features but reduced choroidal neovascularization in mice, suggesting that selectively modulating PPARβ/Γ could be a promising therapeutic approach for treating different forms of AMD.
You can read the full article at https://pmc.ncbi.nlm.nih.gov/articles/PMC5076447/.
Capozzi ME, McCollum GW, Savage SR, Penn JS. Peroxisomeproliferator-activated receptor-β/Γ regulates angiogenic cell behaviors and oxygen-induced retinopathy. Invest Ophthalmol Vis Sci. 2013;54(6):4197-4207. Published 2013 Jun 19. doi:10.1167/iovs.13-11608.
Peroxisomeproliferator-activated receptor-β/Γ regulates angiogenic cell behaviors and oxygen-induced retinopathy
This study shows that activation of PPAR-β/Γ promotes, while its inhibition reduces, preretinal neovascularization (NV) in models of ocular disease. In human retinal endothelial cells, the PPAR-β/Γ agonist GW0742 increased Angptl4 expression and enhanced tube formation, while the antagonist GSK0660 suppressed both proliferation and tube formation. In a rat model of oxygen-induced retinopathy, GW0742 elevated retinal Angptl4 levels and worsened NV, whereas GSK0660 significantly reduced NV. These findings suggest that PPAR-β/Γ contributes to pathological ocular NV and that its inhibition may offer a novel therapeutic strategy.
You can read the full article at https://pmc.ncbi.nlm.nih.gov/articles/PMC3687964/.
At the age of 60, I look and feel better than I ever have in my entire life! Switching my health program and hormone replacement therapy regimen over to Genemedics was one of the best decisions Iāve ever made in my life! Genemedics and Dr George have significantly improved my quality of life and also dramatically improved my overall health. I hav...
Nick Cassavetes ,60 yrs old
Movie Director (āThe Notebookā, āJohn Qā, āAlpha Dogā), Actor and Writer
I am now in my mid-sixties and feel better than I did in my 20ās. Many people have commented that I actually look 20 years younger since I started the program at Genemedics.
Calling Dr. George has proven to be one of the best decisions I have made in my life. Doctors and society convince us that developing various health issues and negative sy...
Pamela Hill ,66 yrs old
Call 800-277-4041 for a Free Consultation
GenemedicsĀ® Health Institute is a global premier institute dedicated to revolutionizing health and medicine through healthy lifestyle education, guidance and accountability in harmony with functional medicine. Our physician-supervised health programs are personally customized to help you reach your health and fitness goals while looking and feeling better than ever.
© Copyright Genemedics Health Institute. All Rights Reserved. Privacy Policy.
STEPS AWAY FROM A YOUNGER. HEALTHIER YOU!
Call 800-277-4041 for a Free Consultation
What to expect during your consultation: