GENEMEDICS APP
GENEMEDICS NUTRITION
Author: Dr. George Shanlikian, M.D. | Last Updated: January 31st, 2024
Bremelanotide offers a wide range of potential benefits, including improving sexual health, promoting weight loss, and enhancing cognitive health. It also contributes to improved blood sugar levels, a better lipid profile, mood enhancement, and increased energy levels. Additionally, it exhibits positive effects on eye health, aids in alcohol addiction treatment, and boosts immune function, making it a versatile compound with multiple potential advantages.
Bremelanotide (BRE mel AN oh tide), also known as PT-141, is a powerful peptide (a compound consisting of two or more amino acids) that activates internal pathways in the brain involved in normal sexual responses. This FDA-approved drug is also a melanocortin receptor agonist, which helps activate a type of protein known as melanocortin receptors. It is given by subcutaneous injection (under the skin) using a single-use autoinjector pen. Scientific studies report that the administration of this peptide has shown promise in various aspects of health.
As a melanocortin receptor agonist, this medication helps activate a type of protein known as melanocortin receptors. These receptors are involved in a wide range of brain activities such as sexual
arousal, mood, appetite, and thinking.
IMG
IMG
Sexual dysfunction, especially low sexual desire, can significantly influence one’s self-confidence and relationship with a sexual partner. Thankfully, for those grappling with low sexual desire, treatments like bremelanotide offer hope. In June 2019, the FDA gave its nod to bremelanotide as a remedy for women experiencing low sexual desire. This approval came after the drug showed an ability to boost sexual desire and lessen distress in two phase 3 trials with women diagnosed with hypoactive sexual desire disorder (HSDD), indicating a clear link between bremelanotide and heightened sexual desire. This makes it the second drug, after flibanserin, to receive such approval by the FDA for treating low sexual desire in premenopausal women, making it a promising solution for those facing challenges in their sexual activity.
Studies on bremelanotide have repeatedly highlighted its potential in addressing low sexual desire and improving overall sexual health:
Studies show that the melanocortin receptor agonist bremelanotide may also help improve body composition by inducing weight loss:
Ready to take control of your weight loss journey? Discover the power of peptides for weight loss and unlock your potential for a healthier, happier you! Explore our comprehensive guide and product offerings today to kickstart your transformation.
There is increasing evidence that bremelanotide along with other melanocortin receptor agonists may help improve cognitive health. Studies show that they exert this effect through the following mechanisms:
Studies show that melanocortin receptor agonists help protect against diabetes and the negative effects of high blood sugar:
There is also evidence that melanocortin receptor agonists such as bremelanotide may help correct abnormal lipid levels:
In men and women with sexual dysfunction, low mood, and depression are very common. Because of the therapeutic benefit of bremelanotide on libido and sexual performance, this powerful peptide has been studied for its mood-enhancing effect:
Photoreceptor cell, a specialized type of neuron found in the retina, is particularly vulnerable to high blood sugar levels. Studies show that melanocortin receptor agonists help preserve eye health:
Recent data have implicated that melanocortin receptor agonists such as bremelanotide may play a role in modulating voluntary ethanol (alcohol) consumption, thereby treating alcohol addiction:
Melanocortin receptor agonists such as bremelanotide play a crucial role in strengthening the immune system. Studies show that melanocortin receptor agonists exert their immune-boosting effects through various mechanisms:
Bremelanotide 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 bremelanotide. However, these side effects weren’t confirmed to be associated with the treatment and could have been a coincidence and not related to the use of bremelanotide. Despite this, it was listed as a side effect associated with bremelanotide. Other side effects not listed may also occur in some patients.
Side effects associated with bremelanotide may include the following:
The dosage of bremelanotide (Vyleesi) is 1.75 mg injected subcutaneously (under the skin) at least 45 minutes before anticipated sexual activity. Do not administer more than 1 dose per 24 hours. Do not use more than 8 doses per month. It’s crucial to follow your healthcare provider’s instructions regarding the appropriate dosage and administration schedule.
Dr. George Shanlikian, renowned as the world’s best hormone therapy doctor, possesses expertise in various medical domains. These include Bio-Identical Hormone Replacement Therapy, Peptide Replacement Therapy, Anti-Aging Medicine, Regenerative Medicine, Stress Management, Nutrition Consulting, Nutritional Supplement Consulting, and Exercise Consulting.
Read more about him here: https://www.genemedics.com/dr-george-shanlikian-md-best-hormone-therapy-doctor
Read more success stories here:
Men’s Success Stories: https://www.genemedics.com/about-ghi/ghi-success-stories/mens-success-stories/
Women’s Success Stories: https://www.genemedics.com/about-ghi/ghi-success-stories/womens-success-stories/
Bremelanotide (BRE mel AN oh tide) is used to treat hypoactive sexual desire disorder (HSDD) in premenopausal women, and it’s an option for erectile dysfunction (ED) in men. It has been shown to improve sexual function and enhance sexual performance.
Bremelanotide is prescribed for women who have not experienced low sexual desire in the past, have not undergone menopause, and continue to have persistently low sexual desire regardless of sexual activity type, context, or partner. It should not be used for treating low sexual desire arising from relationship problems, health concerns, mental disorders, or the use of specific drugs or medications.
Yes, bremelanotide is FDA-approved for the treatment of hypoactive sexual desire disorder in premenopausal women and is considered a viable treatment option.
The brand name for bremelanotide is Vyleesi, and it’s used to address sexual health issues like hypoactive sexual desire disorder and erectile dysfunction (ED).
Yes, men can use bremelanotide for erectile dysfunction treatment. It may also help improve sexual performance and desire.
It is administered as a bremelanotide injection, either as a subcutaneous shot in the abdomen or thigh.
The recommended dosage of bremelanotide varies but is typically a 1.75 mg dose, administered as needed at least 45 minutes before anticipated sexual activity.
Yes. These include nausea, flushing, headache, skin changes, decrease in heart rate, bremelanotide injection site reactions, and high blood pressure (temporary increase in BP).
Common side effects of bremelanotide include nausea, headache, flushing, and high blood pressure. In a comprehensive analysis of two phase 3 trials, investigating the safety and efficacy of a daily 1.75 mg dosage through double-blind, placebo-controlled methods, nausea emerged as a prevalent side effect, typically arising 30 minutes after administration and lasting around 2.4 hours on average. [17] Among participants, seven patients treated with bremelanotide reported 10 treatment-related serious side effects, while four serious adverse events were reported by three placebo recipients. Notably, most serious side effects were transient and characterized as mild to moderate in intensity.
Bremelanotide is considered safe, but it may have potentially serious side effects and drug interactions. It is recommended to watch out for any temporary increase in blood pressure, decrease in heart rate, or severe nausea while using the medication. Most importantly, tell your doctor if you have any medical condition that can affect the treatment or are taking other drugs.
Bremelanotide is not recommended for use during pregnancy, as it might affect the unborn baby. Talk to your doctor if you are planning to use bremelanotide or other medicines.
Bremelanotide typically starts working within 45 minutes, enhancing sexual arousal and desire in the past few minutes after administration.
Bremelanotide activates melanocortin receptors in the brain, leading to increased sexual desire and arousal. Its action is linked to melanocortin receptor agonists.
Certain medications, dietary supplements, and alcohol may interact with bremelanotide, so it’s important to tell your doctor or pharmacist.
Bremelanotide is used to treat erectile dysfunction (ED) and improve sexual performance in men.
Bremelanotide’s effectiveness can vary among individuals, but clinical studies have shown significant increases in sexual desire and arousal.
Yes, bremelanotide can be used to treat erectile dysfunction and other male sexual disorders.
Bremelanotide can increase sexual desire (libido) by influencing brain pathways related to sexual motivation and behavior, potentially enhancing sexual experiences.
Yes, a prescription from a healthcare provider or healthcare professional is required for bremelanotide.
Bremelanotide focuses on increasing sexual desire, different from other medications for ED, some of which primarily affect blood flow.
Combining bremelanotide with other ED medications or other drugs may have potential interactions. Tell your doctor or pharmacist about your current medications before taking bremelanotide.
The success rate of bremelanotide in treating HSDD varies, but studies have shown positive outcomes, especially in women living with this condition. The medication is usually effective in women who have not experienced low sexual desire in the past, have not undergone menopause, and continue to have persistently low sexual desire regardless of sexual activity type, context, or partner.
The bremelanotide autoinjector is used for administering the medication as a subcutaneous injection. Call your doctor if you have any questions regarding bremelanotide injection.
You should inform your healthcare provider about your medical problems (e.g. high blood pressure or uncontrolled hypertension), current medications, any allergies or allergic reactions, and existing conditions, including cardiovascular disease or kidney disease.
Bremelanotide is primarily indicated for premenopausal women with hypoactive sexual desire disorder (HSDD). Its effectiveness in postmenopausal women may vary.
Bremelanotide is primarily indicated for premenopausal women with hypoactive sexual desire disorder (HSDD). Its effectiveness in postmenopausal women may vary.
Bremelanotide addresses hypoactive sexual desire disorder (HSDD) in women, aiming to improve sexual health and sexual function.
Bremelanotide’s increase in sexual desire is believed to be related to its action on specific brain receptors and its influence on sexual motivation and behavior.
Some individuals may experience mental health problems, like mood changes or anxiety, as psychological side effects while using bremelanotide. Call your doctor or contact a poison control center if you experience a mental health problem or if you think you have taken too much of this medicine.
Bremelanotide’s action on melanocortin receptors is linked to dopamine release, associated with pleasure and reward.
Bremelanotide should be stored in a refrigerator between 36°F to 46°F (2°C to 8°C). It should not be frozen and must be kept away from light.
If bremelanotide is not working as expected, call your doctor or consult your healthcare provider for medical attention and guidance.
Individuals with heart conditions or cardiovascular disease may not be good candidates for bremelanotide treatment. Talk to your doctor before using bremelanotide if you have any disease or illness.
Bremelanotide is not a hormone; it works through known mechanisms to enhance desire, different from aphrodisiacs.
Yes, there are age restrictions for using bremelanotide. It is not approved for use in people under the age of 18. This is because the safety and efficacy of bremelanotide in people under 18 have not been established.
Bremelanotide’s safety profile is comparable to other sexual health medications, but individual reactions can vary, including potentially serious side effects. Make sure to talk to your doctor if you have high blood pressure or an unusual or allergic reaction to bremelanotide or other medicines.
Bremelanotide is primarily used for hypoactive sexual desire disorder (HSDD) and erectile dysfunction (ED), but its potential applications in sexual medicine are being explored.
If you miss a dose of bremelanotide, take it as soon as you remember. Only one dose should be taken at a time.
Bremelanotide can be used alongside therapy or counseling for sexual disorders, but coordination with a health care professional is essential.
There might not be a generic version of this medication. It would be advisable to consult with a pharmacist or health care professional for updated information.
This medication is intended for medical use and should not be used recreationally. Misuse may lead to unwanted side effects or interactions.
For more information about bremelanotide injection, consult your healthcare provider, pharmacist, or check reputable medical websites, including official product websites.
Bremelanotide requires a prescription by a healthcare professional and cannot be purchased over the counter.
Several clinical trials have been conducted on bremelanotide to assess its safety and efficacy for hypoactive sexual desire disorder (HSDD) and erectile dysfunction (ED).
Bremelanotide’s availability may vary by country and region. Consultation with a local pharmacist or health care professional can provide specific information.
Bremelanotide is typically supplied as a pre-filled autoinjector for subcutaneous administration.
Bremelanotide: New Drug Approved for Treating Hypoactive Sexual Desire Disorder
Bremelanotide, a newly approved therapy for hypoactive sexual desire disorder (HSDD), was
reviewed through a literature search encompassing relevant studies from January 1996 to December
2019. This search included phase 2 and 3 trials of bremelanotide, resulting in the inclusion of
2 phase 3 and 2 phase 2 trial reports. The data synthesis revealed that bremelanotide
significantly improves desire and reduces distress associated with the lack of desire in
individuals with HSDD. Common adverse effects include nausea, facial flushing, and headache. As
the second FDA-approved treatment for HSDD, bremelanotide’s exact place in therapy is uncertain
due to outdated HSDD guidelines from 2017. While the trials demonstrated statistical
significance in improving sexual desire and related distress, the actual clinical benefit might
be modest. Bremelanotide is administered as a subcutaneous injection around 45 minutes before
sexual activity, deemed safe with minimal drug interactions and no significant interactions with
ethanol. Prescribing guidelines suggest a maximum of 1 dose within 24 hours and no more than 8
doses per month. If no benefit is observed after 8 weeks, discontinuation is recommended.
You can read the abstract at https://pubmed.ncbi.nlm.nih.gov/31893927/
Bremelanotide for Female Sexual Dysfunctions in Premenopausal Women: A Randomized,
Placebo-Controlled Dose-Finding Trial
The study aimed to assess the effectiveness and safety of bremelanotide (BMT), a
melanocortin-receptor-4 agonist, in treating female sexual dysfunctions in premenopausal women.
The patients were divided into groups receiving placebo or different doses of BMT (0.75, 1.25,
or 1.75 mg) via self-administered subcutaneous injections over 12 weeks. The main measurement
was the change in the frequency of satisfying sexual events per month. Secondary measurements
included changes in scores on the female sexual function index and female sexual distress scale,
particularly related to desire, arousal, and orgasm.
The results showed that for the pooled 1.25 mg and 1.75 mg BMT doses compared to placebo, there
were positive changes in the frequency of satisfying sexual events (+0.7 vs. +0.2 events/month),
improvements in the female sexual function index total score (+3.6 vs. +1.9), and reductions in
the female sexual distress scale scores related to desire, arousal, and orgasm (-11.1 vs. -6.8).
Adverse events such as nausea, flushing, and headaches were reported.
In conclusion, the study demonstrated that self-administered subcutaneous BMT was effective,
safe, and well-tolerated in premenopausal women with female sexual dysfunctions. The trial’s
registration number is NCT01382719.
You can read the full article at https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5384512/
Evaluation of the safety and efficacy of bremelanotide, a melanocortin receptor agonist,
in female subjects with arousal disorder: a double-blind placebo-controlled, fixed dose,
randomized study
The article mentioned has been retracted as per the Elsevier Policy on Article Withdrawal. The
retraction was initiated by the Editor-in-Chief of the journal. Following the retraction of Dr.
Safarinejad’s previous work by other journals, The Journal of Sexual Medicine conducted a
comprehensive re-review of all papers authored by Dr. Safarinejad that were published in their
journal. After a thorough reassessment and careful examination of the manuscripts, expert
reviewers identified multiple concerning issues related to the methodology, results, and
statistical interpretation presented in this specific article.
In an effort to address the raised concerns, Dr. Safarinejad was contacted to provide the
original data and explanations. However, Dr. Safarinejad opted not to respond to these
inquiries. Consequently, the journal is unable to verify the accuracy of the results and methods
as presented in the article. As a result, the article has been retracted.
You can read the abstract at https://pubmed.ncbi.nlm.nih.gov/18179455/
An effect on the subjective sexual response in premenopausal women with sexual arousal
disorder by bremelanotide (PT-141), a melanocortin receptor agonist
Hypoactive sexual desire disorder (HSDD) is characterized by a persistent lack of sexual
fantasies and desire, causing distress or difficulties in relationships. Symptoms include
reduced interest in sexual activity, decreased response to sexual cues, loss of interest during
sex, and avoidance of sexual situations. HSDD’s underlying causes involve hormonal,
neurotransmitter, and brain anatomy factors. Its treatment is complex, aiming to address both
biological and psychosocial aspects. Bremelanotide, a melanocortin receptor agonist, has gained
FDA approval for HSDD treatment. Administered intranasally or as a subcutaneous injection, the
recommended dose is 1.75 mg at least 45 minutes before sexual activity. Studies have
demonstrated improved desire, arousal, and orgasm scores with this dosage compared to a placebo,
making bremelanotide a promising option for HSDD treatment.
You can read the full article at https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8788464/
You can read the full article at Female Sexual Dysfunction: Therapeutic Options and
Experimental Challenges
Female sexual dysfunction (FSD) affects around 40% of women and is more common as they age. It
encompasses issues like vaginal dryness, pain during intercourse, reduced arousal, and
difficulty reaching orgasm. Despite its prevalence, few treatment options exist, and only a
minority of affected women seek medical help. Unlike the extensive research and treatment for male erectile dysfunction, FSD has received less attention and is mainly treated through psychological therapy. Cardiovascular diseases like atherosclerosis, peripheral arterial disease, and hypertension, often associated with aging and male erectile dysfunction, have also been linked to FSD. Animal studies have expanded our understanding, but more research is needed to effectively treat FSD. This review aims to provide up-to-date information on FSD, recent progress in understanding its underlying mechanisms, and emerging therapeutic possibilities. You can read the full article at
Designing drugs for the treatment of female sexual dysfunction
About 30% of women experience dysfunction in sexual desire, arousal, or achieving orgasm.
Initial efforts to address female sexual dysfunction were adapted from treatments for male
erectile dysfunction, but these attempts have had limited success. A new approach is emerging,
focusing on targets that potentially influence central pathways involved in sexual function.
Neurotransmitter systems like dopamine and serotonin, along with the neuropeptide melanocortin,
are receiving considerable attention in this context. Preliminary clinical data are encouraging;
however, clinical trial methodologies for female sexual dysfunction are not well-established.
Further testing is necessary to determine whether these treatments can meet regulatory
requirements and effectively address the needs of patients.
You can read the full article at https://www.sciencedirect.com/science/article/abs/pii/S1359644607002759?via%3Dihub
Female Sexual Dysfunction
Female sexual dysfunction significantly impacts the quality of life for numerous women. In the
United States, about 40% of women experience sexual issues, yet these complaints often go
underrecognized and undertreated. The terminology and classification systems for female sexual
dysfunction can be complex, hindering accurate clinical diagnosis. Treatment options for these
dysfunctions are limited, though some interventions show potential benefits and are discussed.
Ongoing research is exploring additional treatments. Establishing clear clinical categories and
diagnostic guidelines for female sexual dysfunction is crucial, as it can greatly aid clinical
practice, public health efforts, and research related to these conditions.
You can read the full article at https://www.sciencedirect.com/science/article/abs/pii/S0025712519300112?via%3Dihub
PT-141: a melanocortin agonist for the treatment of sexual dysfunction
PT-141 is a synthetic peptide similar to alpha-MSH that acts as an agonist at melanocortin
receptors, particularly MC3R and MC4R, primarily located in the central nervous system. In
experiments with rats and nonhuman primates, PT-141 induces penile erections. In rats, systemic
PT-141 administration activates neurons in the hypothalamus, as evidenced by increased c-Fos
immunoreactivity. Neurons in this brain region also demonstrate uptake of pseudorabies virus
injected into the rat penis. When given to both healthy men and those with erectile dysfunction,
PT-141 leads to a rapid, dose-dependent increase in erectile activity. These findings suggest
that PT-141 could be a promising novel treatment for sexual dysfunction.
You can read the abstract at https://nyaspubs.onlinelibrary.wiley.com/doi/abs/10.1111/j.1749-6632.2003.tb03167.x?sid=nlm%3Apubmed
The study evaluated PT-141, a cyclic heptapeptide melanocortin analog, through subcutaneous
administration in healthy males and patients with erectile dysfunction (ED) who had an
inadequate response to Viagra. Inadequate response was defined as achieving suitable erections
for vaginal penetration less than or equal to 50% of the time with 100 mg Viagra. Healthy
subjects received doses from 0.3 to 10 mg, showing a significant erectile response at doses over
1.0 mg. ED patients were given placebo, 4 mg, or 6 mg of PT-141 in a crossover design during
visual sexual stimulation (VSS). Both PT-141 doses induced a statistically significant erectile
response. The treatment was well tolerated and safe in both studies. PT-141’s potential to
induce erections, its tolerability, and its efficacy in patients with inadequate responses to
PDE5 inhibitors suggest it could offer an alternative ED treatment for a broad range of
patients.
You can read the abstract at https://www.nature.com/articles/3901200
The study aimed to assess the safety and effectiveness of intranasal bremelanotide in men
with erectile dysfunction (ED) who didn’t respond to sildenafil, a commonly used ED
medication. The researchers conducted a randomized trial involving 342 married men aged 28
to 59 years with ED. These men were divided into two groups: one receiving 10 mg
bremelanotide as an intranasal spray before sexual activity, and the other receiving a
placebo spray. Participants were required to use the treatment at least 16 times at home.
They were evaluated using medical history, self-administered questionnaires, and assessment
of various ED-related factors.
The results showed that 33.5% of the men in the bremelanotide group experienced positive
clinical outcomes, compared to only 8.5% in the placebo group. Additionally, the
bremelanotide group reported higher satisfaction with sexual intercourse compared to the
placebo group. However, there were more drug-related side effects in the bremelanotide
group. The study concluded that bremelanotide could be a potential alternative treatment for
ED in individuals who do not respond to sildenafil. However, further research is needed to
determine the drug’s effectiveness at different dosages and treatment schedules.
You can read the abstract at https://www.auajournals.org/doi/10.1016/j.juro.2007.10.063..
Melanocortin Receptors, Melanotropic Peptides and Penile Erection
Penile erection is a complex process involving interactions between the nervous system and
specialized vascular tissues. Activation of certain melanocortinergic (MC) receptors in the
central nervous system (CNS) by natural or synthetic melanotropic ligands can trigger or
facilitate spontaneous penile erection. The specific MC receptors involved, mainly MC3 and
MC4 subtypes, have generated conflicting research regarding their roles in erection
initiation. While MC4R seems to be the primary contributor to MC-induced erections, MC3R’s
role remains less understood.
Recent studies involving receptor-specific agonists, antagonists, and knockout mice have shed
light on the distinct contributions of these receptor subtypes. Emerging research suggests
that blocking MC3R in the forebrain might enhance melanocortin-induced erections.
Additionally, melanocortin compounds might interact with established systems like
oxytocinergic pathways at various CNS levels.
Traditional treatments for erectile dysfunction focus on the vascular tissue of the genital
area. Manipulating MC receptors presents an alternative approach, targeting central
mechanisms for treating not only erectile dysfunction but also related sexual dysfunctions
such as decreased sexual motivation and libido loss. The “superpotent” MC agonist PT-141,
derived from MT-II, has progressed to phase II human trials. Melanocortin agonists show
promise as centrally acting therapies, addressing both erectile dysfunction and broader
sexual health concerns.
You can read the full article at https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2694735/
Melanocortins in the treatment of male and female sexual dysfunction
In the past two decades, research on male and female sexual responses has resulted in various
pharmacological options for treating male erectile dysfunction (ED) and female arousal and
orgasmic disorders. Some approaches target peripheral mechanisms like nitric oxide/cyclic
GMP signaling, which affects genital vascular and nonvascular smooth muscles. Others
consider central pathways involved in arousal, orgasmic functions in females, and induction
of penile erection in males. Alongside dopaminergic agonists, drugs affecting the central
melanocortin system are considered promising for addressing sexual dysfunctions in both
genders.
This review outlines progress in the clinical development of melanocortin receptor (MCR)
agonists (melanotan I, melanotan II, bremelanotide) for treating sexual arousal and orgasmic
disorders in adult females, as well as erectile dysfunction in males. The available data
have enhanced our understanding of how the melanocortin pathway regulates male and female
sexual functions. However, more research is needed to fully explore the effects of specific
MCR agonists on penile erection and female arousal and orgasmic functions.
You can read the abstract at https://www.tandfonline.com/doi/abs/10.1517/13543784.2014.934805?journalCode=ieid20
Co-administration of low doses of intranasal PT-141, a melanocortin receptor agonist,
and sildenafil to men with erectile dysfunction results in an enhanced erectile
response
The study aimed to assess the safety and effects of combining subtherapeutic doses of PT-141,
a melanocortin analogue, with sildenafil in patients with erectile dysfunction. Nineteen
patients, who responded to either Viagra or Levitra, underwent a randomized crossover design
involving three different treatments: 25 mg sildenafil and 7.5 mg intranasal PT-141, 25 mg
sildenafil and an intranasal placebo spray, and a placebo tablet and an intranasal placebo
spray. Erectile responses were measured using RigiScan during a 6-hour period after
treatment.
The results demonstrated that co-administration of PT-141 and sildenafil led to a
significantly greater erectile response compared to sildenafil alone. The combination
treatment was found to be safe and well-tolerated, with no new adverse events or increased
frequency/severity of adverse events compared to monotherapy. The findings suggest that
combining intranasal PT-141 with a phosphodiesterase type 5 inhibitor like sildenafil could
be a viable treatment option for patients who do not respond well to higher doses of a
single therapy or have issues with tolerability.
You can read the abstract at https://www.goldjournal.net/article/S0090-4295(04)01272-5/fulltext
The future is today: emerging drugs for the treatment of erectile dysfunction. Expert
opinion on emerging drugs
Erectile dysfunction (ED) is a prevalent male sexual issue affecting 10-20% of men, with
phosphodiesterase type 5 inhibitors (PDE5Is) being the primary treatment. However, there’s a
subset of ED patients not responding to PDE5Is. This review explores emerging compounds for
ED treatment, focusing on both central (clavulanic acid, dopamine, and melanocortin receptor
agonists) and peripheral (novel PDE5Is, guanylate cyclase activators, rho-kinase inhibitors,
and maxi-K channel openers) regulation of penile erection.
The review provides insights into the pathophysiology of erectile function, discussing
mechanisms of action, effectiveness, and adverse effects of various compounds in development
for ED treatment. The emerging drugs are expected to offer personalized treatment,
considering each patient’s unique needs and balancing efficacy with potential side effects.
Combination therapies involving these emerging drugs might be used to enhance effectiveness,
particularly for challenging cases.
You can read the full article at https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3163612/
The Patient Experience of Premenopausal Women Treated with Bremelanotide for
Hypoactive Sexual Desire Disorder: RECONNECT Exit Study Results
Hypoactive sexual desire disorder (HSDD) significantly affects women’s well-being and
relationships. The RECONNECT clinical trials revealed that bremelanotide led to substantial
improvements in sexual desire and related distress in premenopausal women with HSDD,
compared to placebo. Exit surveys and patient interviews were conducted to understand HSDD’s
impact and bremelanotide’s effects from the patients’ viewpoint.
After completing the double-blind study but before entering the open-label extension, around
250 participants were invited to complete an exit survey (17 questions), and up to 90 were
interviewed by phone (17 questions). Patient blinding to the study drug was maintained until
surveys and interviews were finished.
Survey results showed that women receiving bremelanotide experienced heightened sexual
desire, physical arousal, and overall sexual quality in their relationships. In contrast,
placebo recipients reported benefits that didn’t involve the physiological changes described
by the bremelanotide group. Women on bremelanotide reported better communication with their
sexual partner and positive experiences seeking treatment for HSDD.
These exit surveys and interviews confirmed the primary RECONNECT trial findings and offered
a comprehensive understanding of HSDD’s impact on quality of life and patients’ perceptions
of bremelanotide’s effects.
You can read the abstract at https://www.liebertpub.com/doi/10.1089/jwh.2020.8460?url_ver=Z39.88-2003&rfr_id=ori%3Arid%3Acrossref.org&rfr_dat=cr_pub++0pubmed.
Bremelanotide for the Treatment of Hypoactive Sexual Desire Disorder: Two Randomized
Phase 3 Trials
The study aimed to assess the safety and effectiveness of bremelanotide for treating
hypoactive sexual desire disorder (HSDD) in premenopausal women
Two identical phase 3 trials, collectively referred to as RECONNECT, were conducted. These
were randomized, double-blind, placebo-controlled, multicenter trials. Premenopausal women
with HSDD were randomized to receive either bremelanotide 1.75 mg subcutaneously or placebo
for 24 weeks. The primary efficacy endpoints were changes in the Female Sexual Function
Index-desire domain score and Female Sexual Distress Scale-Desire/Arousal/Orgasm item 13
from baseline to the end of the study.
The trials included 1,247 participants in the safety population and 1,202 participants in the
efficacy population. Participants taking bremelanotide experienced statistically significant
increases in sexual desire and reductions in distress related to low sexual desire compared
to those on placebo. The most common side effects associated with bremelanotide were nausea,
flushing, and headache. However, the majority of adverse events were mild or moderate in
intensity.
In conclusion, both studies demonstrated that bremelanotide significantly improved sexual
desire and related distress in premenopausal women with HSDD. The treatment was generally
well-tolerated, with the main adverse events being related to tolerability and mostly mild
or moderate in nature.
You can read the full article at https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6819021/
Long-Term Safety and Efficacy of Bremelanotide for Hypoactive Sexual Desire Disorder
The study aimed to assess the long-term safety and effectiveness of bremelanotide for
treating hypoactive sexual desire disorder (HSDD) in premenopausal women.
Participants who completed the 24-week double-blind core phase of the RECONNECT study, which
consisted of two parallel phase 3 trials (301 and 302), could join a 52-week open-label
extension. This extension evaluated the continuation of bremelanotide treatment for HSDD.
Only those who did not experience serious adverse events during the core phase were eligible
for the extension. Efficacy was measured using the same endpoints as in the core phase, and
adverse events were monitored throughout the extension.
The open-label extension began in 2015 and concluded in 2017. Of the eligible participants,
684 out of 856 chose to participate in the extension, and 272 completed it. The most common
treatment-related adverse events included nausea, flushing, and headache. Notably, no new
safety issues emerged during the extension. Participants who received bremelanotide during
the core phase and continued in the extension maintained improvements in HSDD symptoms, as
measured by the Female Sexual Function Index-desire domain score and Female Sexual Distress
Scale-Desire/Arousal/Orgasm item 13.
In conclusion, the 52-week open-label extension of the RECONNECT study showed sustained
benefits in HSDD symptoms for premenopausal women treated with bremelanotide. The
treatment’s safety profile remained consistent over the extended period, with no new
concerns arising.
You can read the abstract of this article at https://pubmed.ncbi.nlm.nih.gov/31599847/.
Long-Term Safety and Efficacy of Bremelanotide for Hypoactive Sexual Desire
Disorder
The study aimed to assess the long-term safety and efficacy of bremelanotide as a treatment
for hypoactive sexual desire disorder (HSDD) in premenopausal women.
Women who completed the 24-week double-blind core phase of the RECONNECT study, which
consisted of two parallel phase 3 trials (301 and 302) comparing bremelanotide with placebo
in premenopausal women with HSDD, were eligible to participate in a 52-week open-label
extension phase, provided they did not experience serious adverse events during the core
phase. Efficacy was evaluated using the same primary endpoints as in the core phase, and
adverse events were recorded during the extension. Statistical analyses were descriptive in
nature.
The open-label extension for study 301 took place from July 17, 2015, to July 13, 2017, and
for study 302, it occurred from October 5, 2015, to June 29, 2017. Out of the 856 patients
who completed the core phase, 684 chose to participate in the extension, with 272 completing
it. The most commonly reported treatment-related adverse events were nausea (40.4%),
flushing (20.6%), and headache (12.0%). Nausea was the only severe treatment-related adverse
event experienced by multiple participants in both studies during the open-label extension.
The change in Female Sexual Function Index-desire domain score and Female Sexual Distress
Scale-Desire/Arousal/Orgasm item 13 from baseline to the end of the extension ranged from
1.25 to 1.30 and -1.4 to -1.7, respectively, for patients who received bremelanotide during
the core phase. For patients who received placebo during the core phase, the changes were
0.70-0.77 and -0.9, respectively.
In conclusion, the 52-week open-label extension of the RECONNECT study demonstrated sustained
improvements in HSDD symptoms for premenopausal women treated with bremelanotide. No new
safety concerns emerged during this extension period.
You can read the full article at https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6819023/.
Bremelanotide for Treatment of Female Hypoactive Sexual Desire
Hypoactive sexual desire disorder (HSDD) is characterized by persistent lack of sexual
fantasies and desire leading to distress or relationship difficulties. It includes
diminished interest and responsiveness to sexual cues, avoidance of sexual situations, and
reduced motivation for sexual activity. HSDD’s origin involves hormones, neurotransmitters,
and brain structure. Due to its complexity, treatment should address both biological and
psychological aspects.
Bremelanotide, a melanocortin receptor agonist, has received FDA approval for HSDD treatment.
Administered intranasally or subcutaneously, the recommended dosage is 1.75 mg injected into
the abdomen or thigh at least 45 minutes before sexual activity. Clinical studies
demonstrated improved desire, arousal, and orgasm scores compared to a placebo when 1.75 mg
of bremelanotide was taken before sexual activity. This treatment offers promise for
managing HSDD.
You can read the full article at https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8788464/
Bremelanotide for the Treatment of Hypoactive Sexual Desire Disorder: Two Randomized
Phase 3 Trials
The study aimed to assess the safety and efficacy of bremelanotide for treating hypoactive
sexual desire disorder (HSDD) in premenopausal women.
Two identical phase 3 trials, referred to as RECONNECT, were conducted as randomized,
double-blind, placebo-controlled, multicenter trials. Premenopausal women with HSDD were
given subcutaneous bremelanotide 1.75 mg or placebo as needed for 24 weeks. Key efficacy
endpoints were changes in the Female Sexual Function Index-desire domain score and the
Female Sexual Distress Scale-Desire/Arousal/Orgasm item 13.
The trials were conducted between January 2015 and August 2016. Of the 1,267 women
randomized, 1,247 and 1,202 were part of the safety and efficacy populations, respectively.
Majority were white (85.6%), from the U.S. (96.6%), and had an average age of 39 years.
Bremelanotide-treated women experienced statistically significant increases in sexual desire
and significant reductions in distress related to low sexual desire compared to placebo.
Adverse events such as nausea, flushing, and headache were more common in the bremelanotide
group
In conclusion, both studies demonstrated that bremelanotide significantly improved sexual
desire and related distress in premenopausal women with HSDD. The treatment was generally
well-tolerated, with most adverse events being related to tolerability and mild or moderate
in intensity.
You can read the full article at https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6819021/
Bremelanotide for the Treatment of Hypoactive Sexual Desire Disorder: Two Randomized
Phase 3 Trials
The study aimed to assess the safety and efficacy of bremelanotide for treating hypoactive
sexual desire disorder (HSDD) in premenopausal women
Two identical phase 3 trials, referred to as RECONNECT, were conducted as randomized,
double-blind, placebo-controlled, multicenter trials. Premenopausal women with HSDD were
given subcutaneous bremelanotide 1.75 mg or placebo as needed for 24 weeks. Key efficacy
endpoints were changes in the Female Sexual Function Index-desire domain score and the
Female Sexual Distress Scale-Desire/Arousal/Orgasm item 13.
The trials were conducted between January 2015 and August 2016. Of the 1,267 women
randomized, 1,247 and 1,202 were part of the safety and efficacy populations, respectively.
Majority were white (85.6%), from the U.S. (96.6%), and had an average age of 39 years.
Bremelanotide-treated women experienced statistically significant increases in sexual desire
and significant reductions in distress related to low sexual desire compared to placebo.
Adverse events such as nausea, flushing, and headache were more common in the bremelanotide
group
In conclusion, both studies demonstrated that bremelanotide significantly improved sexual
desire and related distress in premenopausal women with HSDD. The treatment was generally
well-tolerated, with most adverse events being related to tolerability and mild or moderate
in intensity.
You can read the full article at https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6819021/
The neurobiology of bremelanotide for the treatment of hypoactive sexual desire
disorder in premenopausal women
Hypoactive sexual desire disorder (HSDD) is a prevalent female sexual dysfunction, impacting
around 10% of U.S. women. This disorder is linked to imbalances in brain hormone and
neurotransmitter levels, causing reduced excitation, increased inhibition, or a combination
of both. Neurotransmitters like dopamine, norepinephrine, serotonin, and hormones like
estradiol and testosterone play roles in female sexual desire and response. Current HSDD
treatments encompass psychotherapy, flibanserin (a serotonin mixed agonist/antagonist), and
bremelanotide (a melanocortin receptor agonist) – both FDA-approved for premenopausal women.
Bremelanotide activates melanocortin receptors (MCRs), neuropeptides associated with the
excitatory pathway of the female sexual response system. The most relevant subtype is MC4R,
situated primarily in the medial preoptic area (mPOA) of the hypothalamus. This region is
pivotal for female sexual function. Bremelanotide’s mechanism of action involves activating
presynaptic MC4Rs on mPOA neurons, leading to increased dopamine release – an excitatory
neurotransmitter associated with heightened sexual desire.
In summary, this review delves into bremelanotide’s mechanism of action within the context of
treating HSDD. It highlights how bremelanotide’s activation of MC4Rs in the hypothalamus can
potentially enhance sexual desire by boosting dopamine release.
You can read the abstract at https://www.cambridge.org/core/journals/cns-spectrums/article/abs/neurobiology-of-bremelanotide-for-the-treatment-of-hypoactive-sexual-desire-disorder-in-premenopausal-women/CB2718A94BC8F52E562867A0F1689B15.
Bremelanotide for the Treatment of Hypoactive Sexual Desire Disorder: Two Randomized
Phase 3 Trials
The study aimed to assess the safety and efficacy of bremelanotide for treating hypoactive
sexual desire disorder (HSDD) in premenopausal women
Two identical phase 3 trials, referred to as RECONNECT, were conducted as randomized,
double-blind, placebo-controlled, multicenter trials. Premenopausal women with HSDD were
given subcutaneous bremelanotide 1.75 mg or placebo as needed for 24 weeks. Key efficacy
endpoints were changes in the Female Sexual Function Index-desire domain score and the
Female Sexual Distress Scale-Desire/Arousal/Orgasm item 13.
The trials were conducted between January 2015 and August 2016. Of the 1,267 women
randomized, 1,247 and 1,202 were part of the safety and efficacy populations, respectively.
Majority were white (85.6%), from the U.S. (96.6%), and had an average age of 39 years.
Bremelanotide-treated women experienced statistically significant increases in sexual desire
and significant reductions in distress related to low sexual desire compared to placebo.
Adverse events such as nausea, flushing, and headache were more common in the bremelanotide
group.
In conclusion, both studies demonstrated that bremelanotide significantly improved sexual
desire and related distress in premenopausal women with HSDD. The treatment was generally
well-tolerated, with most adverse events being related to tolerability and mild or moderate
in intensity
You can read the full article at https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6819021/.>
The Patient Experience of Premenopausal Women Treated with Bremelanotide for
Hypoactive Sexual Desire Disorder: RECONNECT Exit Study Results
Hypoactive sexual desire disorder (HSDD) has a negative impact on women’s overall health and
relationships. Primary analysis from the RECONNECT trials showed significant improvements in
sexual desire and related distress with bremelanotide compared to placebo in premenopausal
women with HSDD. To gauge patient perspectives, exit surveys and interviews were conducted
after the double-blind phase but before the open-label extension.
Around 250 participants completed quantitative exit surveys, and 80 of them also participated
in qualitative telephone interviews. Participants receiving bremelanotide reported increased
sexual desire, physical arousal, and improved overall sexual quality in their relationships.
In contrast, placebo recipients didn’t report similar physiological responses but
highlighted positive experiences seeking HSDD treatment and enhanced communication with
partners
In conclusion, exit surveys and interviews corroborate the primary RECONNECT findings,
offering both quantitative and qualitative insights into how HSDD affects patients’ quality
of life and their perspectives on bremelanotide’s impact
You can read the abstract at https://www.liebertpub.com/doi/10.1089/jwh.2020.8460?url_ver=Z39.88-2003&rfr_id=ori%3Arid%3Acrossref.org&rfr_dat=cr_pub++0pubmed>
Prespecified and Integrated Subgroup Analyses from the RECONNECT Phase 3 Studies of
Bremelanotide
Hypoactive sexual desire disorder (HSDD) is a common female sexual dysfunction characterized
by reduced desire for sexual activity and distress. Bremelanotide, a melanocortin receptor
agonist approved by the FDA for acquired generalized HSDD in premenopausal women, was
investigated for its efficacy compared to placebo across various subgroups in the phase 3
RECONNECT studies
Patients self-administered bremelanotide or placebo before sexual activity for 24 weeks.
Efficacy was assessed using changes in Female Sexual Function Index desire domain and Female
Sexual Distress Scale-Desire/Arousal/Orgasm Item 13. Among the 1,202 patients in the
integrated and subgroup analyses, bremelanotide yielded significant improvements in desire
and decreased distress related to low desire in age, weight, BMI, and bioavailable
testosterone subgroups. These improvements were consistent, with only a few exceptions
Bremelanotide also led to increased reported sexual desire in patients not using hormonal
contraceptives, with a numerical advantage observed in those using contraceptives.
Regardless of contraceptive use, bremelanotide reduced distress significantly. The treatment
achieved improvements in desire and distress irrespective of decreased arousal, HSDD
duration, and across various subgroups
In conclusion, bremelanotide demonstrated significant improvements in sexual desire and
reduced distress in patients with HSDD across multiple predefined subgroups, with only a few
exceptions.
You can read the abstract at https://www.liebertpub.com/doi/10.1089/jwh.2021.0225?url_ver=Z39.88-2003&rfr_id=ori:rid:crossref.org&rfr_dat=cr_pub%20%200pubmed>
An effect on the subjective sexual response in premenopausal women with sexual
arousal disorder by bremelanotide (PT-141), a melanocortin receptor agonist
The study aimed to assess the effects of a single intranasal dose of bremelanotide on
physiological and subjective measures of sexual arousal and desire in premenopausal women
with sexual arousal disorder.
Bremelanotide is a synthetic peptide melanocortin analog that acts as an agonist at
melanocortin receptors MC3R and MC4R, which are associated with various physiological
responses, including sexual behaviors.
Eighteen premenopausal women with sexual arousal disorder participated in a double-blind
study where they received a single intranasal dose of 20 mg bremelanotide or a matching
placebo. Participants viewed neutral and sexually explicit videos while vaginal pulse
amplitude was monitored using photoplethysmography. Questionnaires evaluated perceptions of
sexual response within 24 hours after treatment.
Results showed that more women reported moderate or high sexual desire following
bremelanotide treatment compared to placebo. There was also a trend towards more positive
responses regarding feelings of genital arousal after bremelanotide. Among women attempting
sexual intercourse within 24 hours, more were satisfied with their level of sexual arousal
following bremelanotide compared to placebo. However, vaginal vasocongestion did not
significantly change during viewing erotic videos after bremelanotide administration
compared to placebo. In conclusion, this preliminary study suggests that bremelanotide might
positively impact desire and arousal in women with sexual arousal disorder, making it a
promising candidate for further investigation in at-home studies.
You can read the full article at https://academic.oup.com/jsm/article-abstract/3/4/628/6882956?redirectedFrom=fulltext&login=false.
Ac-Nle-c[Asp-His-DPhe-Arg-Trp-Lys]-NH2 induces penile erection via brain and spinal
melanocortin receptors
The study aimed to investigate the effects of melanocortin peptides on penile erection and
yawning in male rats. The researchers administered the melanocortin receptor agonist MT-II
through various routes (intracerebroventricular, intrathecal, and intravenous) and observed
its impact. They also used the antagonist SHU-9119 to distinguish between spinal and
supraspinal effects. The researchers found that MT-II induced dose-dependent penile
erections, with yawning observed for certain administration routes. Spinal delivery of MT-II
was more effective in inducing erections than other routes. The study confirmed that MT-II
has central proerectile effects and identified melanocortin receptors in the spinal cord
that can trigger erections independently of higher brain centers. These findings offer new
insights into the pathways involved in penile erection and could potentially lead to
improved for erectile dysfunction.
You can read the abstract at https://www.ibroneuroscience.org/article/S0306-4522(02)00866-7/fulltext.
Chronic treatment with a melanocortin-4 receptor agonist causes weight loss, reduces
insulin resistance, and improves cardiovascular function in diet-induced obese rhesus
macaques
The melanocortin-4 receptor (MC4R) plays a significant role in regulating body weight.
Activation of MC4R leads to weight loss in rodents and is linked to obesity in humans. This
makes MC4R a potential target for obesity treatment. However, previous studies have shown
that MC4R agonists can cause various side effects, including increased heart rate and blood
pressure. In this research, a new and highly selective MC4R agonist (BIM-22493 or RM-493)
was tested on obese nonhuman primates. Treatment with this agonist led to short-term reduced
food intake (35%) and sustained weight loss (13.5%) over 8 weeks. The treated animals also
showed improved glucose tolerance and reduced fat accumulation. Importantly, there were no
temporary increase in blood pressure or heart rate with this new agonist. In contrast,
another MC4R agonist (LY2112688) that had previously caused high blood pressure and heart rate in
humans did so again in this study, along with a modest decrease in food intake. This
research highlights that different melanocortin peptide drugs can have varying levels of
effectiveness and side effects.
You can read the full article at https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3554387/..
Melanocortin-4 receptor agonists for the treatment of obesity
The melanocortin receptor family (MC1-5R) and their natural peptide ligands (alpha, beta,
gamma-MSH, and ACTH) play a role in various behavioral and physiological functions,
including the regulation of food intake and body weight. Studies using rodent models have
demonstrated that melanocortin agonists like MTII reduce food intake and body weight, while
antagonists like SHU9119 and AGRP increase food intake and body weight. Deletion of MC3R or
MC4R in mice leads to obesity, with hyperphagia observed mainly in MC4R-deficient mice.
Similarly, in humans, inactivating mutations of MC4R are found in a portion of obese
individuals. This suggests that MC4R activation might reduce food intake in humans,
prompting the search for selective agonists for obesity treatment.
Efforts to develop MC4R-selective small molecule and peptide agonists have seen limited
success over the past decade. Some small molecule agonists have been identified, but few
have shown in vivo activity. However, several potent and selective peptide agonists have
been shown by various researchers to decrease food intake and body weight in rodent models.
This review focuses on assessing the progress in creating small molecule and peptide MC4R
agonists as potential therapies for obesity.
You can read the abstract at https://www.eurekaselect.com/article/23386..
Melanocortin receptor agonist transiently increases oxygen consumption in
rats
The melanocortin receptor family (MC1-5R) and their natural peptide ligands (alpha, beta,
gamma-MSH, and ACTH) play a role in various behavioral and physiological functions,
including the regulation of food intake and body weight. Studies using rodent models have
demonstrated that melanocortin agonists like MTII reduce food intake and body weight, while
antagonists like SHU9119 and AGRP increase food intake and body weight. Deletion of MC3R or
MC4R in mice leads to obesity, with hyperphagia observed mainly in MC4R-deficient mice.
Similarly, in humans, inactivating mutations of MC4R are found in a portion of obese
individuals. This suggests that MC4R activation might reduce food intake in humans,
prompting the search for selective agonists for obesity treatment.
Efforts to develop MC4R-selective small molecule and peptide agonists have seen limited
success over the past decade. Some small molecule agonists have been identified, but few
have shown in vivo activity. However, several potent and selective peptide agonists have
been shown by various researchers to decrease food intake and body weight in rodent models.
This review focuses on assessing the progress in creating small molecule and peptide MC4R
agonists as potential therapies for obesity.
You can read the abstract at https://journals.lww.com/neuroreport/abstract/2001/12040/melanocortin_receptor_agonist_transiently.20.aspx
Effect of bremelanotide on body weight of obese women: Data from two phase 1
randomized controlled trials
The study aims to investigate the effects of bremelanotide, an agonist of the melanocortin 4
receptor (MC4R), on appetite regulation, caloric intake, and body weight in premenopausal
obese women. Two randomized controlled clinical trials were conducted
In Study A, premenopausal women were either given subcutaneous placebo or bremelanotide three
times daily for 15 days. The bremelanotide group exhibited a significant reduction in body
weight (-1.3 kg) and a decrease in caloric intake (around 400 kcal/day) compared to the
placebo group.
Study B involved a crossover design with different treatment sequences, including once-a-day
and twice-a-day exposure to bremelanotide versus placebo. Subjects receiving twice-daily
bremelanotide showed a substantial reduction in body weight (1.7 kg) and a greater decrease
in caloric intake (398-469 kcal) compared to the placebo group. Overall, these trials
indicate that bremelanotide’s agonistic activity at MC4R appears to effectively reduce
caloric intake and promote weight loss in obese premenopausal women
You can read the full article at https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9314948/.
Central serotonin and melanocortin pathways regulating energy homeostasis
The study focuses on understanding how the interaction between the central serotonergic
(5-HT) system and the melanocortin systems within the hypothalamus affects energy balance
and food intake regulation. The hypothalamus plays a crucial role in coordinating responses
to changes in energy availability. Recent research has investigated how serotonin-related
drugs influence pathways related to leptin responsiveness in the hypothalamus.
Through a combination of functional neuroanatomy, feeding experiments, and electrophysiology
studies in rodents, the researchers discovered that anorectic 5-HT drugs require functional
melanocortin pathways to affect food intake. These drugs activate pro-opiomelanocortin
(POMC) neurons in the arcuate nucleus of the hypothalamus. The study suggests that the
serotonin 2C receptor (5-HT(2C)R) is expressed on POMC neurons and contributes to this
effect. Moreover, the researchers found that the anorectic effects of 5-HT drugs are
diminished when downstream melanocortin 3 and 4 receptors are blocked either
pharmacologically or genetically.
The study proposes a model in which the activation of the melanocortin system follows the
activation of the 5-HT system and is essential for the complete anorectic effects of 5-HT
drugs. This research integrates the central 5-HT system into the broader network of
metabolic signals that converge on melanocortin neurons in the hypothalamus to regulate
energy balance and food intake.
You can read the abstract at https://nyaspubs.onlinelibrary.wiley.com/doi/abs/10.1111/j.1749-6632.2003.tb03177.x?sid=nlm%3Apubmed.
The MC4 receptor mediates alpha-MSH induced release of nucleus accumbens
dopamine
There is compelling evidence of a functional connection between the melanocortin and dopamine
systems. The grooming behavior induced by alpha-Melanocyte Stimulating Hormone (alpha-MSH)
appears to be linked to heightened dopaminergic activity in the striatal regions. This
connection, which can be inhibited by dopamine receptor antagonists, hasn’t been
definitively attributed to melanocortin (MC) receptors until now.
Through microdialysis studies on anesthetized rats, researchers administered alpha-MSH into
the ventral tegmental area. This led to a notable increase in dopamine and DOPAC (a dopamine
metabolite) levels in the nucleus accumbens. Importantly, this rise in dopamine was entirely
blocked when the MC4 receptor-specific antagonist HS131 was administered beforehand. This
suggests that the influence of alpha-MSH on dopamine transmission is likely mediated through
the MC4 receptor.
You can read the abstract at
https://journals.lww.com/neuroreport/abstract/2001/07200/the_mc4_receptor_mediates___msh_induced_release_of.22.aspx.
Interaction between alpha-melanocyte-stimulating hormone and corticotropin-releasing
hormone in the regulation of feeding and hypothalamo-pituitary-adrenal
responses
Both alpha-melanocyte-stimulating hormone and corticotropin-releasing hormone (CRH) play
roles in feeding and neuroendocrine processes. The similarity in function and anatomical
distribution between these neurotransmitter systems raised the idea that CRH might be
involved in mediating the central effects of the melanocortin system.
Using double-labeling in situ hybridization, researchers identified that a subset of CRH
neurons in the paraventricular nucleus of the hypothalamus (PVN) contains the melanocortin-4
receptor (MC4R), primarily in the ventromedial region of the parvicellular PVN (up to 33%).
Injecting the melanocortin agonist MTII into conscious rats led to rapid CRH gene
transcription induction in the PVN. This was accompanied by a dose- and time-dependent
increase in plasma corticosterone levels, peaking 30 minutes after MTII injection. The rise
in plasma corticosterone triggered by MTII was reduced by the MC4R antagonist HS014 and the
CRH receptor antagonist alpha-helical-CRH9-41, both in a dose-dependent manner.
Furthermore, the study evaluated the anorectic effect of MTII on food intake at different
time points after injection. The inhibitory impact of MTII on food intake was partly
reversed by pretreatment with alpha-helical-CRH9-41. Collectively, these findings provide
evidence that CRH operates as a downstream mediator of melanocortin signaling, contributing
to how the central melanocortin system regulates feeding and neuroendocrine responses.
You can read the full article at https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6740604/.
Alpha-melanocyte-stimulating hormone stimulates oxytocin release from the dendrites
of hypothalamic neurons while inhibiting oxytocin release from their terminals in the
neurohypophysis
Both the peptides alpha-melanocyte-stimulating hormone (alpha-MSH) and oxytocin produce
similar behavioral effects when administered centrally. This study aimed to explore the
relationship between alpha-MSH and oxytocin in terms of their effects on neuronal activity
and the release of oxytocin.
The study found that alpha-MSH induces the expression of Fos (a marker of neuronal
activation) in the supraoptic nucleus when administered centrally or locally by
retrodialysis. However, this increase in Fos expression does not correlate with an increase
in the electrophysiological excitation of oxytocin neurons in the supraoptic nucleus. In
fact, central injection of alpha-MSH or specific melanocortin-4 receptor (MC4R) agonists
inhibited the electrical activity of oxytocin neurons recorded in vivo. This was supported
by a decrease in oxytocin secretion into the bloodstream after central alpha-MSH injection.
Interestingly, MC4R ligands, including alpha-MSH, induced significant oxytocin release from
dendrites in isolated supraoptic nuclei. This dendritic oxytocin release appears to be
triggered by changes in intracellular calcium concentration ([Ca2+]i). The study observed
that MC4R ligands induce a transient increase in [Ca2+]i in oxytocin neurons, which could be
due to the mobilization of calcium from intracellular stores rather than entry through
voltage-gated channels.
In summary, this research reveals that alpha-MSH can lead to differential regulation of
dendritic release and systemic secretion of oxytocin. While alpha-MSH induces Fos
expression, it inhibits the electrical activity of oxytocin neurons. However, MC4R ligands
stimulate oxytocin release from dendrites through intracellular calcium mobilization. This
study highlights the complex interplay between these peptides in regulating oxytocin-related
processes.
You can read the full article at https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6741015/.
Melanocortin Receptor Agonists Facilitate Oxytocin-Dependent Partner Preference
Formation in the Prairie Vole
The central melanocortin (MC) system, particularly the MC4 receptor (MC4R), interacts with
neurochemical systems that regulate socioemotional behaviors, including oxytocin (OT) and
dopamine. In monogamous prairie voles, OT and dopamine work together to promote partner
preference formation, a measure of lasting social bonding between mates. This study aimed to
explore how MC receptor activation influences partner preference formation in prairie voles
and the interaction between the MC and OT systems during this process.
Administering the MC3/4R receptor peptide agonist Melanotan II (MTII) and the selective MC4R
agonist Pf-446687 peripherally enhanced partner preference formation in prairie voles, but
not in non-monogamous meadow voles. The partner preferences induced by MTII were enduring,
persisting even a week after drug treatment. The study suggests that the prosocial effects
of MC receptor agonists might involve modulation of the OT system, as coadministration of an
OT receptor antagonist prevented MTII-induced partner preferences.
Furthermore, MTII selectively activated hypothalamic OT neurons and boosted central OT
release. As OT is known to enhance certain aspects of social cognition in humans, the
findings imply that MC4R could potentially be a therapeutic target for enhancing social
function in psychiatric disorders like autism spectrum disorders and schizophrenia, possibly
through the activation of the OT system.
You can read the full article at https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4839509/.
Neonatal melanocortin receptor agonist treatment reduces play fighting and promotes
adult attachment in prairie voles in a sex-dependent manner
The study focuses on the impact of the melanocortin receptor (MCR) system, known for its
involvement in feeding and sexual behavior, on social behavior, which has been less
explored. Specifically, the study examines the long-term effects of early-life MCR
stimulation on social development in prairie voles.
Researchers administered the MC3/4R agonist melanotan-II (MTII) to neonatal male and female
prairie voles between postnatal days 1 and 7. The study found that male voles treated with
MTII during this period showed reduced initiation of play fighting bouts as juveniles
compared to control males. Neonatal exposure to MTII facilitated the formation of partner
preferences in adult females but not males after a brief cohabitation with an opposite-sex
partner.
Additionally, acute injections of MTII triggered significant immediate early gene EGR-1
immunoreactivity in hypothalamic oxytocin, vasopressin, and corticotropin-releasing factor
neurons in neonatal animals. Daily neonatal treatment with a more selective MC4R agonist,
PF44687, led to the development of partner preferences in both adult females and males. In
conclusion, early-life exposure to MCR agonists resulted in persistent changes in social
behavior in prairie voles, suggesting potential structural or functional alterations in the
neural circuits involved in forming social relationships. The study highlights the complex
role of the MCR system in regulating social behavior beyond its better-studied effects on
feeding and sexual behavior.
You can read the full article at https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4158739/
Nucleus accumbens oxytocin and dopamine interact to regulate pair bond formation in
female prairie voles
The study investigates the interactions between oxytocin (OT) and dopamine (DA) systems in
pair bond formation in monogamous prairie voles. While both OT and DA have been linked to
pair bonding, the precise nature of their interactions and the brain circuits involved
remain unexplored.
The study reveals that both OT and DA D2-type receptors are crucial for pair bond formation.
Blocking either type of receptor prevents partner preferences induced by either OT or a
D2-type agonist. The nucleus accumbens (NAcc) is identified as a key brain region for these
interactions. In the NAcc, blocking OT receptors hinders the partner preferences induced by
a D2-type agonist, while blocking D2-type DA receptors (but not D1-type receptors) hampers
OT-induced partner preferences.
In summary, the study concludes that the simultaneous activation of OT and DA D2-type
receptors in the nucleus accumbens is essential for the formation of pair bonds in female
prairie voles. This highlights the intricate interplay between these neurochemical systems
and specific brain regions in regulating pair bonding behaviors.
You can read the abstract at https://www.ibroneuroscience.org/article/S0306-4522(03)00555-4/fulltext.
Oxytocin and the neural mechanisms regulating social cognition and affiliative
behavior
Oxytocin, produced in the hypothalamus and released through the neurohypophyseal system, has
dual roles: peripheral release aids in childbirth and milk ejection, while central release
coordinates maternal nurturing and mother-infant bonding. Recent research highlights
oxytocin’s broader impact on affiliative behavior in both sexes. It influences alloparental
care and pair bonding in female monogamous prairie voles and social recognition in male and
female mice. In humans, oxytocin enhances social behaviors like increased eye gaze towards
faces, interpersonal trust, and the ability to interpret emotions from facial expressions.
Although the neurohypophyseal oxytocin system is well understood, less is known about
oxytocin release within the brain. This review explores oxytocin’s role in regulating
prosocial interactions and delves into the neuroanatomy of the central oxytocin system.
You can read the full article at https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2748133/.
D-cycloserine facilitates socially reinforced learning in an animal model relevant to
autism spectrum disorders
Currently, there are no drugs that specifically address the social challenges of autism
spectrum disorders (ASD). This gap could be attributed to a lack of relevant animal models
and behavioral paradigms for ASD. Partner preference formation in prairie voles represents a
form of socially reinforced learning, making it a valuable model for investigating social
cognitive processes. D-cycloserine (DCS), a cognitive enhancer acting on
N-methyl-D-aspartate receptors, is known to promote learning. The study aimed to explore
whether DCS could enhance socially reinforced learning in the partner preference paradigm,
potentially aiding social functioning in ASD when combined with behavioral therapies.
Female prairie and meadow voles received either peripheral or direct brain region injections
of DCS, including the nucleus accumbens (linked to reinforcement learning) and the amygdala
(associated with social information processing). After cohabiting with a male vole under
non-preference-inducing conditions, the development of a preference for the stimulus male
vole over a novel one was measured through a partner preference test.
Results indicated that a low dose of peripheral DCS enhanced partner preference formation in
prairie voles, but not meadow voles, in situations where it wouldn’t naturally occur. This
effect was reproduced in prairie voles using direct microinfusions of DCS into the nucleus
accumbens and amygdala. In conclusion, partner preference formation in prairie voles offers
a relevant behavioral paradigm for identifying pharmacotherapeutics addressing social
deficits. The study suggests that DCS, when combined with social behavioral therapy, could
be a potential treatment strategy for the social challenges associated with ASD
You can read the full article at https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3164818/
Distinct Effects of Leptin and a Melanocortin Receptor Agonist Injected Into Medial
Hypothalamic Nuclei on Glucose Uptake in Peripheral Tissues
The study aimed to determine the contributions of the leptin receptor and melanocortin
receptors (MCRs) in different medial hypothalamic nuclei to the regulation of peripheral
glucose uptake. Leptin, a hormone influencing various physiological processes, was injected
into specific medial hypothalamic nuclei in mice, and its effects on glucose uptake in
peripheral tissues were observed. Additionally, an MCR agonist, MT-II, was also administered
to examine its impact on glucose uptake.
Results showed that injecting leptin into the ventromedial hypothalamus (VMH) increased
glucose uptake in skeletal muscle, brown adipose tissue (BAT), and the heart. Leptin
injection into the arcuate nucleus (ARC) enhanced glucose uptake in BAT, while injections
into the dorsomedial hypothalamus (DMH) and paraventricular hypothalamus (PVH) had no effect
on glucose uptake. The MCR antagonist SHU9119 blocked the effects of leptin in the VMH.
Administering MT-II into the VMH or intracerebroventricularly increased glucose uptake in
skeletal muscle, BAT, and the heart, while MT-II injection into the PVH increased glucose
uptake in BAT. However, MT-II injections into the DMH or ARC did not influence glucose
uptake.
Conclusions drawn from the study indicate that the VMH plays a role in mediating glucose
uptake in response to both leptin and MT-II in skeletal muscle, BAT, and the heart. These
effects were dependent on MCR activation. The leptin receptor in the ARC and MCR in the PVH,
on the other hand, were found to regulate glucose uptake in BAT. Overall, different medial
hypothalamic nuclei have distinct roles in mediating glucose uptake in response to leptin
and MT-II in peripheral tissues.
You can read the full article at https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2780865/
Analysis of the therapeutic functions of novel melanocortin receptor agonists in
MC3R- and MC4R-deficient C57BL/6J mice
The study focuses on novel non-selective melanocortin receptor agonists (BIM-22493,
BIM-22511) that can cross the blood-brain barrier when administered peripherally. The
effects of these agonists on obesity, insulin sensitivity, and liver metabolism were
investigated in diet-induced obese mice.
When administered peripherally, these melanocortin agonists improved obesity,
hyperinsulinemia, and fatty liver disease in obese mice. The specificity of their function
was confirmed using mice lacking melanocortin-3 receptor (MC3RKO) and melanocortin-4
receptor (MC4RKO). Obesity, hyperinsulinemia, and hepatosteatosis were observed in MC4RKO
mice, but not MC3RKO mice. The improvement in obesity and metabolic parameters associated
with acute BIM-22493 treatment and 14 days of BIM-22511 treatment required functional MC4R,
while MC3R was not essential.
Chronic treatment with BIM-22511 reduced fasting insulin levels even in MC4RKO mice,
demonstrating that the improvement in hyperinsulinemia is partially independent of MC4R.
However, this treatment did not fully resolve hepatosteatosis in MC4RKO mice or influence
hepatic lipogenic gene expression. In summary, peripherally administered melanocortin
receptor agonists exert effects on body weight, liver metabolism, and glucose homeostasis
through distinct pathways. MC4R is crucial for the agonist-induced weight loss and
improvements in liver metabolism, while improvements in hyperinsulinemia can involve other
melanocortin receptors. The study suggests that agonists targeting various melanocortin
receptors could have potential for addressing glucose homeostasis abnormalities linked to
obesity.
You can read the full article at https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2755620/
Serotonin 2C Receptor Agonists Improve Type 2 Diabetes via Melanocortin-4 Receptor
Signaling Pathways
The increasing burden of type 2 diabetes and its associated health issues necessitates new
and effective treatments. This study reveals that serotonin 2C receptor (5-HT(2C)R)
agonists, known for their appetite-suppressing effects, significantly enhance glucose
tolerance and reduce plasma insulin levels in murine models of obesity and type 2 diabetes.
Importantly, these glucose-improving effects were observed at doses that did not impact
eating behavior, energy expenditure, activity levels, body weight, or fat mass. The study
further shows that the beneficial effects on glucose homeostasis are dependent on the
activation of downstream melanocortin-4 receptors (MC4Rs), but not MC3Rs. This suggests that
targeting 5-HT(2C)Rs pharmacologically might provide a novel approach to improving glucose
tolerance in type 2 diabetes independently of weight-related changes.
You can read the full article at https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2075535/
Central melanocortin receptor agonist reduces hepatic lipogenic gene expression in
streptozotocin-induced diabetic mice
The study aimed to investigate whether the central melanocortin system plays a role in
regulating hepatic lipogenic gene expression under insulin-deficient conditions. Using a
mouse model of insulin-deficient diabetes induced by streptozotocin (STZ), the researchers
administered the melanocortin agonist MTII intracerebroventricularly (i.c.v.) to the
diabetic mice. The effects of MTII treatment on hepatic gene expression were examined.
Results showed that MTII treatment reduced the expression levels of genes involved in lipid
biosynthesis, including stearoyl-CoA desaturase 1 (SCD1), glycerol-3-phosphate
acyltransferase 1 (GPAT1), acyl-CoA:diacylglycerol acyltransferase 1 (DGAT1), and DGAT2,
without affecting insulin levels, insulin resistance, or glucose tolerance. MTII treatment
also decreased the protein levels of fatty acid synthase (FAS) and SCD1 in the liver.
Additionally, the expression levels of transcription factors associated with these lipogenic
genes, sterol regulatory element-binding protein 1c (SREBP-1c) and peroxisome
proliferator-activated receptor γ2 (PPARγ2), were reduced by MTII treatment.
These findings suggest that central melanocortin signaling can regulate hepatic lipogenic
gene expression independently of insulin levels. Enhancing central melanocortin signaling
might offer a potential approach to addressing abnormal lipid metabolism associated with
insulin deficiency or insufficiency.
You can read the abstract at https://www.sciencedirect.com/science/article/abs/pii/S0024320511000622?via%3Dihub
Obesity hypertension: role of leptin and sympathetic nervous system
besity is strongly linked to essential hypertension, with excess renal sodium reabsorption
and increased sympathetic activity playing key roles. Leptin, a hormone involved in appetite
regulation, may partially connect obesity and heightened sympathetic activity. Studies
indicate that leptin can increase renal sympathetic activity and elevate blood pressure and
heart rate through adrenergic activation. Leptin’s effects on blood pressure may involve
interactions with neurochemical pathways in the hypothalamus, including melanocortin-4
receptors. However, the exact mechanisms by which leptin influences blood pressure, whether through these
pathways or others like neuropeptide Y, remain uncertain. Leptin has additional actions,
including promoting nitric oxide formation and improving insulin sensitivity, which could
mitigate blood pressure effects in certain conditions. Given the growing obesity epidemic,
further research is needed to fully understand these complex interactions and their
relevance to human obesity-related hypertension.
You can read the full article at https://academic.oup.com/ajh/article/14/S3/103S/205038?login=false
The central melanocortin system directly controls peripheral lipid metabolism
Disruptions in the central nervous melanocortin system (CNS-Mcr) have been linked to obesity. Researchers investigated the role of this system in adiposity control, independent of nutrient intake. Inhibiting melanocortin receptors (Mcr) in rats and disrupting the Mc4r gene in mice led to increased lipid uptake, triglyceride synthesis, and fat accumulation in white adipose tissue (WAT). Conversely, enhanced CNS-Mcr signaling triggered lipid mobilization. These effects were not tied to food intake and occurred before changes in adiposity. Reduced CNS-Mcr signaling also improved insulin sensitivity and glucose uptake in WAT while decreasing glucose utilization in muscle and brown adipose tissue. These changes were linked to the sympathetic nervous system and liver triglyceride synthesis. These findings explain how decreased melanocortin signaling promotes adiposity even without increased eating and demonstrates its influence on substrate utilization. The study also reveals the molecular mechanisms underlying CNS-Mcr control over lipid metabolism, offering insights for designing more effective methods to manage adiposity.
You can read the full article at https://www.ncbi.nlm.nih.gov/pmc/articles/PMC1978426/
Central melanocortin receptors regulate insulin action
Leptin regulates energy balance and insulin action through the melanocortin pathway in the hypothalamus. This study investigated the impact of manipulating the melanocortin pathway on insulin action and adiposity distribution. Rats were infused with a melanocortin receptor agonist (alpha-MSH) or antagonist (SHU9119) while maintaining constant food intake. Alpha-MSH reduced intra-abdominal fat and significantly enhanced insulin’s effects on glucose uptake and production. In contrast, SHU9119 had opposite effects. These findings reveal a neuroendocrine network that plays a crucial role in connecting energy intake with insulin action.
You can read the full article at https://www.ncbi.nlm.nih.gov/pmc/articles/PMC200952/.
Melanocortin receptor agonists MCR1-5 protect photoreceptors from high-glucose damage and restore antioxidant enzymes in primary retinal cell culture
Melanocortin receptors, particularly subtypes 1 and 5, play a protective role in diabetic retinopathy, a condition where retinal photoreceptors are vulnerable to high glucose levels and oxidative stress. This study investigated the effects of these receptors in primary retinal cell cultures exposed to high glucose concentrations. Treatment with agonists for melanocortin receptors 1 and 5 reduced inflammation and enhanced antioxidant enzyme levels, helping to preserve photoreceptor integrity. These findings suggest that these receptors have a significant role in safeguarding primary retinal cells from damage caused by high glucose or oxidative stress.
You can read the full article at https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5387132/
Activation of melanocortin receptors MC1 and MC5 attenuates retinal damage in experimental diabetic retinopathy
In a mouse model of streptozotocin-induced diabetic retinopathy (DR), the study aimed to investigate the potential protective effects of melanocortin receptors (MC). About 80% of the diabetic mice developed microvascular changes resembling DR without vascular leakage. Occludin, a protein important for maintaining blood-retinal barrier integrity, decreased in these mice. Expression of MC receptors Mc1r and Mc5r was observed in the retina. Intravitreal injection of selective MC1 and MC5 agonists resulted in significant protection, maintaining regular retinal vessel structure. Diabetic mice had elevated levels of inflammatory and angiogenic factors in the retina, while MC agonists reversed these changes. MC3-MC4 agonist/antagonists did not affect the assessed parameters. These findings suggest that MC receptors, particularly MC1 and MC5, could activate protective mechanisms against DR-related changes in the retina.
You can read the full article at https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4753692/
Melanocortin 1 receptor: structure, function, and regulation
The melanocortin 1 receptor (MC1R) is a Gs protein-coupled receptor with a pivotal role in regulating skin pigmentation, UV responses, and melanoma risk. It is highly polymorphic, and its loss of function leads to fair skin, UV sensitivity, and higher melanoma susceptibility due to impaired skin pigmentation and DNA repair. MC1R signaling involves adenylyl cyclase activation, generating cAMP as a second messenger. Hormonally controlled by melanocortin, agouti signaling protein, and β-defensin 3, cAMP signaling via MC1R increases melanin production and deposition in the skin’s epidermis. This melanin helps shield against UV penetration and enhances nucleotide excision repair (NER), a genomic stability mechanism that eliminates UV-induced DNA damage to prevent mutagenesis. This review outlines MC1R’s structure, function, and its influence on NER pathways.
You can read the full article at https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4885833/
The protective effects of the melanocortin receptor (MCR) agonist, melanotan-II (MTII), against binge-like ethanol drinking are facilitated by deletion of the MC3 receptor in mice
Recent studies have linked the melanocortin (MC) system to the modulation of voluntary ethanol consumption. Melanotan-II (MTII), a nonselective melanocortin receptor (MCR) agonist, has been shown to reduce voluntary ethanol consumption in mice. Prior research indicated that central administration of MTII reduces ethanol drinking through the MC4R receptor, as observed in mice lacking MC4R expression. However, the neurocircuitry involved in binge-like ethanol drinking differs from moderate consumption. This study aimed to explore the role of MC3R in binge-like ethanol intake using the “drinking in the dark” (DID) procedure, a model for binge-like drinking. MC3R+/+ and MC3R-/- mice were given MTII intracerebroventricularly before a 4-hour ethanol access period. MTII reduced binge-like ethanol drinking in both types of mice, with MC3R-/- mice showing increased sensitivity to the protective effects of MTII. These findings suggest that MC3Rs counteract the protective effects of MTII against binge-like ethanol consumption, implying potential therapeutic use of selective MC3R antagonists in addressing excessive ethanol intake.
You can read the full article at https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3946855/
Evidence that Melanocortin Receptor Agonist Melanotan-II Synergistically Augments the Ability of Naltrexone to Blunt Binge-Like Ethanol Intake in Male C57BL/6J Mice
The study investigates the potential synergy between the melanocortin (MC) receptor agonist melanotan-II (MTII) and the opioid receptor antagonist naltrexone (NAL) in reducing binge-like alcohol (ethanol) consumption in mice. The authors explore the interaction between the MC and opioid systems, both of which modulate behaviors like pain, drug tolerance, and food intake. Using a mouse model of binge drinking, they find that both NAL and MTII independently reduce binge-like ethanol intake and blood ethanol levels. Interestingly, when administered together, a low dose of MTII significantly enhances the effectiveness of NAL in reducing binge-like ethanol consumption, leading to a synergistic effect. These findings suggest that targeting MC signaling, possibly in combination with opioid antagonism, could offer a novel strategy for treating alcohol abuse disorders and enhancing existing naltrexone-based therapies.
You can read the full article at https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4515169/
Effects of Melanocortin Receptor Activation and Blockade on Ethanol Intake: A Possible Role for the Melanocortin-4 Receptor
The study examines the role of melanocortin receptors (MCRs) in modulating ethanol consumption. The researchers investigate the effects of MCR agonists and antagonists on alcohol intake in mice with different genetic backgrounds. They find that the nonselective MCR agonist melanotan II (MTII) effectively reduces ethanol consumption in both Mc3r-deficient and wild-type mice, indicating that the MC3 receptor is not critical for this effect. However, intracerebroventricular infusion of the highly selective MC4R agonist decreases ethanol drinking, while administration of the MCR antagonist AgRP-(83-132) increases ethanol consumption. These results suggest that the MC4 receptor, rather than MC3 receptor, plays a role in MCR agonist-induced reduction of ethanol intake, and that antagonistic actions of AgRP-(83-132) lead to increased ethanol consumption. This indicates that compounds targeting MCRs could hold promise for treating alcohol abuse disorders in addition to obesity.
You can read the full article at https://www.ncbi.nlm.nih.gov/pmc/articles/PMC1360238/
The melanocortin system in leukocyte biology
The review discusses the components of the melanocortin system, including melanocortin peptides, receptors (MCRs), and endogenous antagonists. Melanocortin peptides have various effects on the body, including anti-inflammatory and immunomodulatory effects. Leukocytes both produce and respond to melanocortins, making them a significant target for these peptides. Activation of MCRs by melanocortin ligands reduces inflammation by inhibiting the translocation of the NF-kappaB nuclear factor, which plays a pivotal role in the inflammatory process. The review explores how melanocortin peptides and receptors influence leukocyte biology, highlighting their role in regulating inflammation.
You can read the full article at https://jlb.onlinelibrary.wiley.com/doi/full/10.1189/jlb.0706426
POMC gene-derived peptides activate melanocortin type 3 receptor on murine macrophages, suppress cytokine release, and inhibit neutrophil migration in acute experimental inflammation
This study investigated the effects of ACTH-related peptides in a murine model of experimental gout. Systemic administration of ACTH4-10 and alpha- and beta-melanocyte stimulating hormones inhibited neutrophil accumulation without affecting blood cell counts or corticosterone levels. ACTH4-10 significantly reduced the release of the chemokine KC, a marker of inflammation. In vitro, ACTH and ACTH4-10 inhibited macrophage activation, including phagocytosis and KC release, with IC50 values of approximately 30 nM and 100 µM, respectively. The melanocortin receptor type 3/4 antagonist SHU9119 blocked the inhibitory effects of ACTH4-10 both in vitro and in vivo. Although melanocortin type 3 receptor mRNA was detected in mouse peritoneal macrophages, not type 4 receptor mRNA was found. The study suggests that the activation of melanocortin type 3 receptors by ACTH4-10 attenuates cytokine release from macrophages, providing an anti-inflammatory mechanism for ACTH that is independent of glucocorticoid release.
You can read the abstract at https://pubmed.ncbi.nlm.nih.gov/10358199/
[D-Trp8]-gamma-melanocyte-stimulating hormone exhibits anti-inflammatory efficacy in mice bearing a nonfunctional MC1R (recessive yellow e/e mouse)
The study investigated the anti-inflammatory effects of melanocortin receptors (MC1 and MC3R) using a selective MC3R agonist called [d-Trp(8)]-gamma-melanocyte-stimulating hormone (MSH). Recessive yellow (e/e) mice with nonfunctional MC1R were used. [d-Trp(8)]-gamma-MSH activated MC3R in peritoneal macrophages, reducing cAMP accumulation and chemokine KC release triggered by urate crystals. Synthetic and natural MC3R antagonists, SHU9119 and AGRP, blocked the effect, while the MC4R antagonist HS024 did not. Systemic [d-Trp(8)]-gamma-MSH treatment inhibited KC release and cell accumulation induced by urate crystals. [d-Trp(8)]-gamma-MSH also induced the anti-inflammatory protein heme-oxygenase 1 (HO-1). The study suggests that [d-Trp(8)]-gamma-MSH could be developed for treating inflammatory conditions due to its dual mechanism involving cytokine/chemokine inhibition and HO-1 induction.
You can read the abstract at https://pubmed.ncbi.nlm.nih.gov/16959942/
Inflamed phenotype of the mesenteric microcirculation of melanocortin type 3 receptor-null mice after ischemia-reperfusion. FASEB journal : official publication of the Federation of American Societies for Experimental Biology
This study investigated the role of melanocortin receptor type 3 (MC3R) in vascular inflammation. Using MC3R-null mice, the researchers found that these mice exhibited increased plasma extravasation, cell adhesion, and emigration after occlusion and reopening of the superior mesenteric artery. The MC3R-null mice also showed elevated expression of inflammatory molecules like CCL2, CXCL1, and myeloperoxidase. Both MC1R and MC3R were present in the inflamed mesenteric tissue, but only MC3R had a significant impact on vascular responses. Treating animals with a selective MC3R agonist led to reduced cell adhesion, emigration, and chemokine production, highlighting the inhibitory role of MC3R in regulating cell trafficking and local mediator generation in vascular inflammation.
You can read the full article at https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2700033/
A role for MC3R in modulating lung inflammation
In this study, researchers investigated the potential of melanocortin peptides as therapeutic agents for lung inflammation, both in allergic and non-allergic models. They identified that melanocortin receptor type 1 (MC1R) and type 3 (MC3R) are expressed on alveolar macrophages. Stimulation with the MC3R agonist [D-TRP(8)]-gamma-MSH and pan-agonist alpha-MSH increased cAMP levels in wild-type and recessive yellow (e/e) mice, but not in MC3R-null mice. In allergic and non-allergic inflammation models, alpha-MSH and [D-TRP(8)]-gamma-MSH reduced leukocyte accumulation and inhibited TNF-alpha release. These effects were significantly attenuated in MC3R-null mice, suggesting that MC3R activation plays a key role in mediating the anti-inflammatory effects of melanocortins. The study suggests that selective MC3R agonists could be promising anti-inflammatory agents for lung inflammation therapy.
You can read the full article at https://www.sciencedirect.com/science/article/abs/pii/S1094553908000928?via%3Dihub
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 I’ve 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 (“The Notebook”, “John Q”, “Alpha Dog”), Actor and Writer
I am now in my mid-sixties and feel better than I did in my 20’s. 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
Genemedics® 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: