Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 176
Filter
Add more filters

Publication year range
1.
Am J Pathol ; 194(1): 150-164, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37827217

ABSTRACT

Corneal endothelial cells (CEnCs) regulate corneal hydration and maintain tissue transparency through their barrier and pump function. However, these cells exhibit limited regenerative capacity following injury. Currently, corneal transplantation is the only established therapy for restoring endothelial function, and there are no pharmacologic interventions available for restoring endothelial function. This study investigated the efficacy of the neuropeptide α-melanocyte-stimulating hormone (α-MSH) in promoting endothelial regeneration during the critical window between ocular injury and the onset of endothelial decompensation using an established murine model of injury using transcorneal freezing. Local administration of α-MSH following injury prevented corneal edema and opacity, reduced leukocyte infiltration, and limited CEnC apoptosis while promoting their proliferation. These results suggest that α-MSH has a proregenerative and cytoprotective function on CEnCs and shows promise as a therapy for the prevention and management of corneal endothelial dysfunction.


Subject(s)
Cornea , Corneal Edema , alpha-MSH , Female , Pregnancy , Animals , Mice , Mice, Inbred BALB C , Humans , Cell Line , Cornea/cytology , Endothelial Cells , Corneal Edema/drug therapy , Corneal Edema/pathology , Tissue Preservation , alpha-MSH/therapeutic use , Cytoprotection , Neutrophil Infiltration , Monocytes/metabolism , Macrophages/metabolism , Wound Healing/drug effects
2.
Lasers Surg Med ; 55(5): 490-502, 2023 07.
Article in English | MEDLINE | ID: mdl-37051852

ABSTRACT

OBJECTIVES: One symptom of hypertrophic scar (HTS) that can develop after burn injury is dyschromia with hyper- and hypopigmentation. There are limited treatments for these conditions. Previously, we showed there is no expression of alpha melanocyte stimulating hormone (α-MSH) in hypopigmented scars, and if these melanocytes are treated with synthetic α-MSH in vitro, they respond by repigmenting. The current study tested the same hypothesis in the in vivo environment using laser-assisted drug delivery (LADD). METHODS: HTSs were created in red Duroc pigs. At Day 77 (pre), they were treated with CO2 fractional ablative laser (FLSR). Synthetic α-MSH was delivered as a topical solution dissolved in  l-tyrosine (n = 6, treated). Control scars received LADD of  l-tyrosine only (n = 2, control). Scars were treated and examined weekly through Week 4. Digital images and punch biopsies of hyper, hypo-, and normally pigmented scar and skin were collected. Digital pictures were analyzed with ImageJ by tracing the area of hyperpigmentation. Epidermal sheets were obtained from punch biopsies through dispase separation and RNA was isolated. qRT-PCR was run for melanogenesis-related genes: tyrosinase (TYR), tyrosinase-related protein-1 (TYRP1), and dopachrome tautomerase (DCT). Two-way ANOVA with multiple comparisons and Dunnett's correction compared the groups. RESULTS: The areas of hyperpigmentation were variable before treatment. Therefore, data is represented as fold-change where each scar was normalized to its own pre value. Within the LADD of NDP α-MSH + l-tyrosine group, hyperpigmented areas gradually increased each week, reaching 1.3-fold over pre by Week 4. At each timepoint, area of hyperpigmentation was greater in the treated versus the control (1.04 ± 0.05 vs. 0.89 ± 0.08, 1.21 ± 0.07 vs. 0.98 ± 0.24, 1.21 ± 0.08 vs. 1.04 ± 0.11, 1.28 ± 0.11 vs. 0.94 ± 0.25; fold-change from pre-). Within the treatment group, pretreatment, levels of TYR were decreased -17.76 ± 4.52 below the level of normal skin in hypopigmented scars. After 1 treatment, potentially due to laser fractionation, the levels decreased to -43.49 ± 5.52. After 2, 3, and 4 treatments, there was ever increasing levels of TYR to almost the level of normally pigmented skin (-35.74 ± 15.72, -23.25 ± 6.80, -5.52 ± 2.22 [p < 0.01, Week 4]). This pattern was also observed for TYRP1 (pre = -12.94 ± 1.82, Week 1 = -48.85 ± 13.25 [p < 0.01], Weeks 2, 3, and 4 = -34.45 ± 14.64, -28.19 ± 4.98, -6.93 ± 3.05 [p < 0.01, Week 4]) and DCT (pre = -214.95 ± 89.42, Week 1 = -487.93 ± 126.32 [p < 0.05], Weeks 2, 3, and 4 = -219.06 ± 79.33, -72.91 ± 20.45 [p < 0.001], -76.00 ± 24.26 [p < 0.001]). Similar patterns were observed for scars treated with LADD of  l-tyrosine alone without NDP α-MSH. For each gene, in hyperpigmented scar, levels increased at Week 4 of treatment compared to Week 1 (p < 0.01). CONCLUSIONS: A clinically-relevant FLSR treatment method can be combined with topical delivery of synthetic α-MSH and l-tyrosine to increase the area of pigmentation and expression of melanogenesis genes in hypopigmented HTS. LADD of  l-tyrosine alone leads to increased expression of melanogenesis genes. Future studies will aim to optimize drug delivery, timing, and dosing.


Subject(s)
Cicatrix, Hypertrophic , Hyperpigmentation , Hypopigmentation , Lasers, Gas , Animals , Swine , Cicatrix, Hypertrophic/drug therapy , Cicatrix, Hypertrophic/genetics , Cicatrix, Hypertrophic/pathology , Tyrosine , alpha-MSH/therapeutic use , alpha-MSH/metabolism , Pharmaceutical Preparations , Pigmentation , Hypopigmentation/drug therapy , Hypopigmentation/genetics , Hyperpigmentation/drug therapy , Hyperpigmentation/genetics , Lasers, Gas/therapeutic use , Melanins/metabolism
3.
Int J Mol Sci ; 24(8)2023 Apr 08.
Article in English | MEDLINE | ID: mdl-37108092

ABSTRACT

The endogenous neuropeptide α-Melanocyte Stimulating Hormone (α-MSH) is a potent suppressor of inflammation and has an essential role in maintaining the normal anti-inflammatory microenvironment of the retina. While the therapeutic use of α-MSH peptide in uveitis and diabetic retinopathy models has been demonstrated, its short half-life and instability limit its use as a therapeutic drug. A comparable analog, PL-8331, which has a stronger affinity to melanocortin receptors, longer half-life, and, so far, is functionally identical to α-MSH, has the potential to deliver melanocortin-based therapy. We examined the effects of PL-8331 on two mouse models of retinal disease, Experimental Autoimmune Uveoretinitis (EAU) and Diabetic Retinopathy (DR). PL-8331 therapy applied to mice with EAU suppressed EAU and preserved retinal structures. In diabetic mice, PL-8331 enhanced the survival of retinal cells and suppressed VEGF production in the retina. In addition, retinal pigment epithelial cells (RPE) from PL-8331-treated diabetic mice retained normal anti-inflammatory activity. The results demonstrated that the pan-melanocortin receptor agonist PL-8331 is a potent therapeutic drug to suppress inflammation, prevent retinal degeneration, and preserve the normal anti-inflammatory activity of RPE.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Retinopathy , Uveitis , Mice , Animals , alpha-MSH/pharmacology , alpha-MSH/therapeutic use , Diabetic Retinopathy/drug therapy , Melanocortins , Diabetes Mellitus, Experimental/drug therapy , Retina , Uveitis/drug therapy , Inflammation/drug therapy , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use
4.
Exp Eye Res ; 218: 108986, 2022 05.
Article in English | MEDLINE | ID: mdl-35196505

ABSTRACT

The melanocortin system plays an essential role in the regulation of immune activity. The anti-inflammatory microenvironment of the eye is dependent on the expression of the melanocortin-neuropeptide alpha-melanocyte stimulating hormone (α-MSH). In addition, the melanocortin system may have a role in retinal development and retinal cell survival under conditions of retinal degeneration. We have found that treating experimental autoimmune uveitis (EAU) with α-MSH suppresses retinal inflammation. Also, this augmentation of the melanocortin system promotes immune tolerance and protection of the retinal structure. The benefit of α-MSH-therapy appears to be dependent on different melanocortin receptors. Therefore, we treated EAU mice with α-MSH-analogs with different melanocortin-receptor targets. This approach demonstrated which melanocortin-receptors suppress inflammation, preserve retinal structure, and induce immune tolerance in uveitis. At the chronic stage of EAU the mice were injected twice 1 day apart with 50 µg of α-MSH or an α-MSH-analog. The α-MSH-analogs were a pan-agonist PL8331, PL8177 (potent MC1r-only agonist), PL5000 (a pan-agonist with no MC5r functional activity), MT-II (same as PL5000) and PG901 (MC5r agonist, but also an antagonist to MC3r, and MC4r). Clinical EAU scores were measured until resolution in the α-MSH-treated mice, when the eyes were collected for histology, and spleen cells collected for retinal-antigen-stimulated cytokine production. Significant suppression of EAU was seen with α-MSH or PL8331 treatment. This was accompanied with significant preservation of retinal structure. A similar effect was seen in EAU-mice that were treated with PL8177, except the suppression of EAU was temporary. In EAU mice treated with PL5000, MTII, or PG901, there was no suppression of EAU with a significant loss in whole retina and outer-nuclear layer thickness. There was significant suppression of IL-17 with induction of IL-10 by retinal-antigen stimulated spleen T cells from EAU mice treated with α-MSH, PL8331, PL8177, or PL5000, but not from EAU mice treated with MT-II, or PG901. Our previous studies show the melanocortin system's importance in maintaining ocular immune privilege and that α-MSH-treatment accelerates recovery and induces retinal-antigen-specific regulatory immunity in EAU. Our current results show that this activity is centered around MC1r and MC5r. In addition, the results suggest that a therapeutic potential to target MC1r and MC5r together to suppress uveitis induces regulatory immunity with potentially maintaining a normal retinal structure.


Subject(s)
Uveitis , alpha-MSH , Animals , Inflammation/metabolism , Mice , Receptors, Melanocortin/metabolism , Retina/metabolism , Uveitis/metabolism , alpha-MSH/pharmacology , alpha-MSH/therapeutic use
5.
Neurochem Res ; 47(2): 394-408, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34586586

ABSTRACT

Persons with type 1 diabetes have an increased risk of stroke compared with the general population. α-Melanocyte-stimulating hormone (α-MSH) is a neuropeptide that has protective effects against ischemia/reperfusion (I/R) induced organ damages. In this study, we aimed to investigate the neuroprotective role of this peptide on I/R induced brain damage after experimental stroke associated with hyperglycemia using C57BL/6J Ins2Akita/+ mice. Experimental stroke was induced by blocking the right middle cerebral artery for 2 h with reperfusion for 2 and 22 h, respectively using the intraluminal method. Animals were treated intraperitoneally with or without α-MSH at 1 h after ischemia and 1 h after reperfusion. Significantly higher survival rate and lower neurological scores were recorded in animals injected with α-MSH. Similarly, neuron death, glial cells activation as well as oxidative and nitrosative stress were significantly decreased in α-MSH treated group. Relative intensities of matrix metallopeptidases 9, cyclooxygenase 2 and nuclear factor-κB were significantly decreased while intensities of Akt, heme oxygenase (HO) 1, HO-2 and B-cell lymphoma 2 were significantly increased after α-MSH treatment. In addition, gene expressions of monocarboxylate transporter (MCT) 1, MCT-2 and activity-regulated cytoskeleton-associated protein were significantly higher in brain samples treated with α-MSH, suggesting this peptide may have role in neuron survival by an involvement of lactate metabolism. In conclusion, α-MSH is neuroprotective under hyperglycemic condition against I/R induced brain damage by its anti-inflammatory, anti-oxidative and anti-apoptotic properties. The use of α-MSH analogues may be potential therapeutic agents for diabetic stroke.


Subject(s)
Brain Ischemia , Diabetes Mellitus, Type 1 , Reperfusion Injury , Animals , Brain Ischemia/drug therapy , Diabetes Mellitus, Type 1/drug therapy , Humans , Mice , Mice, Inbred C57BL , Reperfusion Injury/metabolism , alpha-MSH/pharmacology , alpha-MSH/therapeutic use
6.
Diabetes Obes Metab ; 24(4): 583-598, 2022 04.
Article in English | MEDLINE | ID: mdl-34882941

ABSTRACT

The melanocortin-4 receptor (MC4R) has been critically investigated for the past two decades, and novel findings regarding MC4R signalling and its potential exploitation in weight loss therapy have lately been emphasized. An association between MC4R and obesity is well established, with disease-causing mutations affecting 1% to 6% of obese patients. More than 200 MC4R variants have been reported, although conflicting results as to their effects have been found in different cohorts. Most notably, some MC4R gain-of-function variants seem to rescue obesity and related complications via specific pathways such as beta-arrestin (ß-arrestin) recruitment. Broadly speaking, however, dysfunctional MC4R dysregulates satiety and induces hyperphagia. The picture at the mechanistic level is complicated as, in addition to the canonical G stimulatory pathway, the ß-arrestin signalling pathway and ions (particularly calcium) seem to interact with MC4R signalling to contribute to or alleviate obesity pathogenesis. Thus, the overall complexity of the MC4R signalling spectra has broadened considerably, indicating there is great potential for the development of new drugs to manage obesity and its related complications. Alpha-melanocyte-stimulating hormone is the major endogenous MC4R agonist, but structure-based ligand discovery studies have identified possible superior and selective agonists that can improve MC4R function. However, some of these agonists characterized in vitro and in vivo confer adverse effects in patients, as demonstrated in clinical trials. In this review, we provide a comprehensive insight into the genetics, function and regulation of MC4R and its contribution to obesity. We also outline new approaches in drug development and emerging drug candidates to treat obesity.


Subject(s)
Obesity , Receptor, Melanocortin, Type 4 , Drug Development , Homeostasis , Humans , Obesity/drug therapy , Obesity/genetics , Obesity/metabolism , Receptor, Melanocortin, Type 4/genetics , Receptor, Melanocortin, Type 4/metabolism , alpha-MSH/metabolism , alpha-MSH/pharmacology , alpha-MSH/therapeutic use
7.
CNS Spectr ; 27(3): 281-289, 2022 06.
Article in English | MEDLINE | ID: mdl-33455598

ABSTRACT

Hypoactive sexual desire disorder (HSDD) is a common female sexual dysfunction and is estimated to affect approximately 10% of women in the United States. It has been suggested that HSDD is associated with an imbalance of hormone and neurotransmitter levels in the brain, resulting in decreased excitation, increased inhibition, or a combination of both. Evidence suggests neurotransmitters, including dopamine (DA), norepinephrine, and serotonin, as well as hormones such as estradiol and testosterone, contribute to female sexual desire and response. Current treatments for HSDD include psychotherapy, and two US Food and Drug Administration-approved medications for premenopausal women: flibanserin, a serotonin mixed agonist and antagonist, and bremelanotide, a melanocortin receptor (MCR) agonist. Melanocortins are endogenous neuropeptides associated with the excitatory pathway of the female sexual response system. MCRs are found throughout the body, including the brain. Bremelanotide is an MCR agonist that nonselectively activates several of the receptor subtypes, of which subtype 4 (MC4R) is the most relevant at therapeutic doses. MC4R is predominantly expressed in the medial preoptic area (mPOA) of the hypothalamus in the brain, and is important for female sexual function. Animal studies suggest that bremelanotide may affect female sexual desire by activating presynaptic MC4Rs on neurons in the mPOA of the hypothalamus, leading to increased release of DA, an excitatory neurotransmitter that increases sexual desire. This review presents what is known about the mechanism of action of bremelanotide in the context of treating HSDD.


Subject(s)
Serotonin , Sexual Dysfunctions, Psychological , Animals , Dopamine/metabolism , Female , Humans , Neurotransmitter Agents/therapeutic use , Peptides, Cyclic , Serotonin/metabolism , Sexual Dysfunctions, Psychological/drug therapy , alpha-MSH/therapeutic use
8.
Lung ; 200(4): 463-472, 2022 08.
Article in English | MEDLINE | ID: mdl-35717488

ABSTRACT

PURPOSE: Alpha-melanocyte stimulating hormone (α-MSH) is known to have anti-inflammatory effects. However, the anti-inflammatory properties of α-MSH on normal bronchial epithelial cells are largely unknown, especially in the context of in vitro sarcoidosis models. METHODS: We evaluated the anti-inflammatory effects of α-MSH on two different in vitro sarcoidosis models (lung-on-membrane model; LOMM and three-dimensional biochip pulmonary sarcoidosis model; 3D-BSGM) generated from NBECs and an in vivo sarcoidosis mouse model. RESULTS: Treatment with α-MSH decreased inflammatory cytokine levels and downregulated type I interferon pathway genes and related proteins in LOMM and 3D-BSGM models. Treatment with α-MSH also significantly decreased macrophages and cytotoxic T-cells counts in a sarcoidosis mice model. CONCLUSION: Our results confirm the direct role of type I IFNs in the pathogenesis of sarcoid lung granulomas and highlight α-MSH as a potential novel therapeutic agent for treating pulmonary sarcoidosis.


Subject(s)
Sarcoidosis, Pulmonary , Sarcoidosis , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Granuloma/drug therapy , Inflammation/metabolism , Mice , Sarcoidosis/drug therapy , Sarcoidosis, Pulmonary/drug therapy , alpha-MSH/metabolism , alpha-MSH/pharmacology , alpha-MSH/therapeutic use
9.
Exp Eye Res ; 210: 108685, 2021 09.
Article in English | MEDLINE | ID: mdl-34252414

ABSTRACT

Dry eye (DE) is a chronic, multifactorial ocular surface disease associated with visual disturbance, tear film instability, hyperosmolarity, ocular surface inflammation and damage. Effective intervention is necessary to control this disease. In this study we topically applied α-melanocyte stimulating hormone (α-MSH) on the ocular surface of scopolamine-induced DE rats and found that it promoted tear secretion, reduced tear breakup time and fluorescein sodium staining and increased the number of conjunctival goblet cells. To investigate the mechanism, protein array was conducted, which showed that α-MSH exerted its effects via epithelial growth factor receptor (EGFR) in the JAK-STAT signaling pathway. Furthermore, in vitro experiments showed that α-MSH protected human corneal epithelial cells (hCECs) by maintaining their migration ability and viability and decreasing apoptosis. However, blockade of EGFR abolished these protective effects. Moreover, α-MSH decreased the level of autophagy in benzalkonium chloride (BAC)-stressed hCECs via EGFR. These results demonstrated that α-MSH ameliorated lesions and restored ocular surface functions by upregulating EGFR expression.


Subject(s)
Dry Eye Syndromes/drug therapy , ErbB Receptors/genetics , Gene Expression Regulation/physiology , Hormones/therapeutic use , alpha-MSH/therapeutic use , Administration, Ophthalmic , Animals , Apoptosis , Autophagy , Cell Line , Cell Movement/physiology , Cell Survival/physiology , Disease Models, Animal , Dry Eye Syndromes/chemically induced , Dry Eye Syndromes/genetics , Dry Eye Syndromes/pathology , Epithelium, Corneal/drug effects , Epithelium, Corneal/metabolism , Epithelium, Corneal/pathology , Female , Flow Cytometry , Goblet Cells/drug effects , Hormones/administration & dosage , Humans , Ophthalmic Solutions , RNA Interference , Rats , Rats, Wistar , Scopolamine/toxicity , Tears/physiology , alpha-MSH/administration & dosage
10.
Photochem Photobiol Sci ; 20(2): 315-320, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33721252

ABSTRACT

BACKGROUND: Afamelanotide (AFA) is a synthetic analogue of α-melanocyte-stimulating hormone that is approved for the treatment of patients affected by erythropoietic protoporphyria (EPP). AFA induces a "sun free" tanning and changes of acquired melanocytic nevi (AMN) that are generically described as "darkening". OBJECTIVES: To assess clinical and dermoscopic AMN changes during AFA treatment. METHODS: Adult EPP patients treated with two AFA implants 50 days apart were enrolled. They underwent a clinical and dermoscopic examination of all AMN at baseline (T0), and after 5 (T1) and 12 (T2) months from the first AFA implant. The general pattern, symmetry, number, and size of pigmented globules, morphology of the pigment network, and dermoscopic melanoma features were assessed. RESULTS: Fifteen patients were enrolled with 103 AMN. At T1 all reticular and 2-component AMN showed a focal network thickening that returned to baseline by T2. The increase of globules' number was observed at T1 but not at T2. The difference in number was not influenced by patients' age or phototype. Dermoscopic changes suggestive of malignancy were never seen. The development of new AMN was never registered. CONCLUSIONS: AFA treatment induces reversible changes of AMN dermoscopic morphology without findings suggestive of malignant transformation and it does not stimulate the development of new AMN.


Subject(s)
Dermatologic Agents/adverse effects , Nevus, Pigmented/diagnosis , Protoporphyria, Erythropoietic/pathology , alpha-MSH/analogs & derivatives , Adult , Dermatologic Agents/therapeutic use , Dermoscopy , Female , Humans , Male , Middle Aged , Nevus, Pigmented/etiology , Protoporphyria, Erythropoietic/drug therapy , Receptor, Melanocortin, Type 1/metabolism , Sunlight , Time Factors , alpha-MSH/adverse effects , alpha-MSH/therapeutic use
11.
Dermatology ; 237(6): 995-999, 2021.
Article in English | MEDLINE | ID: mdl-34464955

ABSTRACT

BACKGROUND: Melanotan II (MT II) is a synthetic analogue of α-melanocyte-stimulating hormone that, via interaction with the melanocortin 1 receptor, induces skin hyperpigmentation. The unregulated acquisition of MT II injections via the internet and other outlets has become popular over the last decades in order to exploit its properties for use as a tanning agent. Due to the covert nature of MT II use, it is difficult to assess the extent of its use among the general population and to characterise any associated side effects. OBJECTIVES: The aim of this study was to qualitatively examine MT II use, as portrayed on online forums, and to explore the motivations for its use and side effect profile. METHODS: Data were extracted retrospectively from UK and Ireland online chatrooms and forums from January 2016 to October 2017. Inclusion criteria were active MT II chatrooms and forums considered to be within the public domain. An inductive thematic analysis identified themes within discussion threads. RESULTS: A total of 623 discussion entries were extracted; 205 participants contributed to these entries. Emergent themes included motivation for MT II use, misinformation in the context of using an unregulated product, product preparation and administration, dosing regimens, sunbed use, side effects and concerning practices associated with MT II use. CONCLUSION: Motivations for MT II use included the pursuit of a tanned appearance, often in anticipation of sun holidays and fitness/body building competitions. Clinicians should be aware not only of the potential risks in relation to pigmented skin lesions, but also remain cognisant of the other medical hazards associated with the use of this substance, namely transmission of infectious diseases, use of potentially contaminated products, polypharmacy, and sunbed exposure.


Subject(s)
Internet , Motivation , Peptides, Cyclic/therapeutic use , Skin Pigmentation/drug effects , alpha-MSH/analogs & derivatives , Humans , Patient Satisfaction , Qualitative Research , Retrospective Studies , Sunbathing , United Kingdom , alpha-MSH/therapeutic use
12.
Clin Exp Dermatol ; 46(7): 1189-1204, 2021 Oct.
Article in English | MEDLINE | ID: mdl-33872410

ABSTRACT

Systemic photoprotection aims to negate the negative effects of ultraviolet radiation-induced DNA damage. Systemic supplements may be used as a monotherapy or in combination with topical sunscreens. Using the keywords 'carotenoids', 'flavonoids', 'systemic photoprotection', 'polyphenols' and 'polypodium leucotomos extract', we searched the databases MEDLINE and EMBASE to find relevant English-language articles. Few trials have supported the use of any of these supplements as monotherapy, impeding the recommendation of these systemic supplements as an alternative to sunscreen for photoprotection. Nicotinamide has exhibited clinically relevant benefits in reducing nonmelanoma skin cancers in trials and could be recommended as an adjunctive therapy for the most vulnerable indviduals. Further research is required, which needs to be of higher statistical power, using more clinically meaningful outcome measures with comparison to the current gold standard of care (topical photoprotection) to support the use of alternative therapies in clinical practice.


Subject(s)
Dietary Supplements , Plant Extracts/therapeutic use , Skin Neoplasms/prevention & control , Sunscreening Agents , Antioxidants/therapeutic use , Carotenoids/therapeutic use , Dermatologic Agents/therapeutic use , Flavonoids/therapeutic use , Humans , Ultraviolet Rays , alpha-MSH/analogs & derivatives , alpha-MSH/therapeutic use
13.
Clin Exp Dermatol ; 46(2): 248-258, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33350506

ABSTRACT

Vitiligo is an autoimmune skin condition characterized by depigmented macules and patches, and has a huge psychosocial impact on patients. Treatment of vitiligo aims to prevent the spread of disease and facilitate repigmentation of affected lesions. The mainstay of treatment for unstable vitiligo has been topical agents (corticosteroids, calcineurin inhibitors) and phototherapy. However, systemic treatments are increasingly being shown to have a significant impact on the course of the disease as monotherapy or adjunctive therapy. Of note, oral mini-pulsed corticosteroid therapy, methotrexate, minocycline, ciclosporin, Janus kinase inhibitors and certain supplements have been used in the systemic treatment of vitiligo. We review the underlying evidence supporting the use of each of these systemic treatments.


Subject(s)
Adrenal Cortex Hormones/therapeutic use , Calcineurin Inhibitors/therapeutic use , Neoadjuvant Therapy/methods , Phototherapy/methods , Vitiligo/pathology , Vitiligo/therapy , Administration, Oral , Administration, Topical , Adrenal Cortex Hormones/administration & dosage , Adrenal Cortex Hormones/adverse effects , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/adverse effects , Anti-Bacterial Agents/therapeutic use , Calcineurin Inhibitors/administration & dosage , Calcineurin Inhibitors/adverse effects , Combined Modality Therapy , Cyclosporine/administration & dosage , Cyclosporine/adverse effects , Cyclosporine/therapeutic use , Dermatologic Agents/administration & dosage , Dermatologic Agents/adverse effects , Dermatologic Agents/therapeutic use , Humans , Janus Kinase Inhibitors/administration & dosage , Janus Kinase Inhibitors/adverse effects , Janus Kinase Inhibitors/therapeutic use , Methotrexate/administration & dosage , Methotrexate/adverse effects , Methotrexate/therapeutic use , Minocycline/administration & dosage , Minocycline/adverse effects , Minocycline/therapeutic use , Outcome Assessment, Health Care , Phototherapy/adverse effects , Psychology , Vitiligo/psychology , alpha-MSH/administration & dosage , alpha-MSH/adverse effects , alpha-MSH/analogs & derivatives , alpha-MSH/therapeutic use
14.
Int J Mol Sci ; 22(9)2021 Apr 25.
Article in English | MEDLINE | ID: mdl-33922961

ABSTRACT

Leptin and its receptor are essential for regulating food intake, energy expenditure, glucose homeostasis and fertility. Mutations within leptin or the leptin receptor cause early-onset obesity and hyperphagia, as described in human and animal models. The effect of both heterozygous and homozygous variants is much more investigated than compound heterozygous ones. Recently, we discovered a spontaneous compound heterozygous mutation within the leptin receptor, resulting in a considerably more obese phenotype than described for the homozygous leptin receptor deficient mice. Accordingly, we focus on compound heterozygous mutations of the leptin receptor and their effects on health, as well as possible therapy options in human and animal models in this review.


Subject(s)
Obesity/therapy , Receptors, Leptin/genetics , Receptors, Leptin/metabolism , Animals , Anti-Obesity Agents/therapeutic use , Bariatric Surgery , Disease Models, Animal , Heterozygote , Humans , Hyperphagia/genetics , Mice , Mutation , Obesity/genetics , Receptors, Leptin/chemistry , alpha-MSH/analogs & derivatives , alpha-MSH/therapeutic use
15.
Diabetes Obes Metab ; 22(11): 2133-2140, 2020 11.
Article in English | MEDLINE | ID: mdl-32627316

ABSTRACT

AIM: To report an analysis of ~1 year of setmelanotide treatment for obesity and hunger, as well as metabolic and cardiac outcomes, in individuals with Bardet-Biedl syndrome (BBS). MATERIALS AND METHODS: Individuals aged 12 years and older with BBS received once-daily setmelanotide. The dose was titrated every 2 weeks to establish the individual therapeutic dose (≤3 mg); treatment continued for an additional 10 weeks. Participants who lost 5 kg or more (or ≥5% of body weight if <100 kg at baseline) continued into the 52-week extension phase. The primary outcome was mean percent change from baseline in body weight at 3 months. Hunger scores and safety were secondary outcomes. RESULTS: From February 2017 and February 2018, 10 individuals were screened; eight completed the 3-month treatment phase and seven completed the extension phase. Mean percent change in body weight from baseline to 3 months was -5.5% (90% CI, -9.3% to -1.6%; n = 8); change from baseline was -11.3% (90% CI, -15.5% to -7.0%; n = 8) at 6 months and -16.3% (90% CI, -19.9% to -12.8%; n = 7) at 12 months. All participants reported at least one treatment-emergent adverse event (AE), most commonly injection-site reaction. No AEs led to study withdrawal or death. Most, morning, and average hunger scores were reduced across time points. CONCLUSIONS: Setmelanotide reduced body weight and hunger in individuals with BBS and had a safety profile consistent with previous reports. Setmelanotide may be a treatment option in individuals with BBS-associated obesity and hyperphagia.


Subject(s)
Bardet-Biedl Syndrome , Receptor, Melanocortin, Type 4 , Bardet-Biedl Syndrome/drug therapy , Bardet-Biedl Syndrome/epidemiology , Humans , Obesity/complications , Obesity/drug therapy , alpha-MSH/analogs & derivatives , alpha-MSH/therapeutic use
16.
Ann Pharmacother ; 54(7): 684-690, 2020 07.
Article in English | MEDLINE | ID: mdl-31893927

ABSTRACT

Objective: To review data regarding bremelanotide, a recently approved therapy for hypoactive sexual desire disorder (HSDD). Data Sources: Literature search of Medline, SCOPUS, and EMBASE was performed using the search terms bremelanotide, bremelanotide injection, Vyleesi, and melanocortin 4 receptor agonist between January 1, 1996, and December 15, 2019. Reference lists from included articles were also reviewed for pertinent citations. Study Selection/Data Extraction: We included phase 2 and 3 trials of bremelanotide. There were 2 reports of phase 3 trials and 2 reports of phase 2 trials. Additional information from supplementary analyses was also referenced. Data Synthesis: Bremelanotide demonstrates significant improvement in desire and a significant decrease in distress related to lack of desire. The most common adverse effects include nausea (39.9%), facial flushing (20.4%), and headache (11%). Relevance to Patient Care and Clinical Practice: Bremelanotide is the second Food and Drug Administration-approved medication for the treatment of HSDD. Bremelanotide's place in therapy is unknown, as the HSDD guidelines were last updated in 2017. Although the trials met statistical significance for change in sexual desire elements and distress related to sexual desire, the clinical benefit may only be modest. Conclusion: Bremelanotide is a subcutaneous injection that can be administered as needed approximately 45 minutes prior to sexual activity. Bremelanotide is safe and has limited drug-drug interactions, including no clinically significant interactions with ethanol. Prescribing guidelines recommend no more than 1 dose in 24 hours and no more than 8 doses per month. Individuals should discontinue use after 8 weeks without benefit.


Subject(s)
Libido/drug effects , Peptides, Cyclic/therapeutic use , Sexual Dysfunctions, Psychological/drug therapy , alpha-MSH/therapeutic use , Clinical Trials as Topic , Drug Interactions , Female , Humans , Injections, Subcutaneous , Nausea/chemically induced , Peptides, Cyclic/administration & dosage , Peptides, Cyclic/adverse effects , Peptides, Cyclic/pharmacokinetics , Receptor, Melanocortin, Type 3/agonists , Receptor, Melanocortin, Type 4/agonists , Treatment Outcome , alpha-MSH/administration & dosage , alpha-MSH/adverse effects , alpha-MSH/pharmacokinetics
18.
J Hepatol ; 71(2): 422-433, 2019 08.
Article in English | MEDLINE | ID: mdl-31102718

ABSTRACT

Porphyrias are rare inherited disorders caused by specific enzyme dysfunctions in the haem synthesis pathway, which result in abnormal accumulation of specific pathway intermediates. The symptoms depend upon the chemical characteristics of these substances. Porphyrins are photoreactive and cause photocutaneous lesions on sunlight-exposed areas, whereas accumulation of porphyrin precursors is related to acute neurovisceral attacks. Current therapies are suboptimal and mostly address symptoms rather than underlying disease mechanisms. Advances in the understanding of the molecular bases and pathogenesis of porphyrias have paved the way for the development of new therapeutic strategies. In this Clinical Trial Watch we summarise the basic principles of these emerging approaches and what is currently known about their application to porphyrias of hepatic origin or with hepatic involvement.


Subject(s)
Acetylgalactosamine/analogs & derivatives , Bone Marrow Transplantation/methods , Cholestyramine Resin/therapeutic use , Genetic Therapy/methods , Liver Transplantation/methods , Porphyrias, Hepatic/drug therapy , Porphyrias, Hepatic/surgery , Pyrrolidines/therapeutic use , Receptor, Melanocortin, Type 1/agonists , alpha-MSH/analogs & derivatives , 5-Aminolevulinate Synthetase/antagonists & inhibitors , Acetylgalactosamine/pharmacology , Acetylgalactosamine/therapeutic use , Heme/biosynthesis , Humans , Liver/metabolism , Porphyrias, Hepatic/classification , Porphyrias, Hepatic/pathology , Porphyrins/metabolism , Pyrrolidines/pharmacology , alpha-MSH/therapeutic use
19.
N Engl J Med ; 375(3): 240-6, 2016 Jul 21.
Article in English | MEDLINE | ID: mdl-27468060

ABSTRACT

Patients with rare defects in the gene encoding proopiomelanocortin (POMC) have extreme early-onset obesity, hyperphagia, hypopigmentation, and hypocortisolism, resulting from the lack of the proopiomelanocortin-derived peptides melanocyte-stimulating hormone and corticotropin. In such patients, adrenal insufficiency must be treated with hydrocortisone early in life. No effective pharmacologic treatments have been available for the hyperphagia and obesity that characterize the condition. In this investigator-initiated, open-label study, two patients with proopiomelanocortin deficiency were treated with setmelanotide, a new melanocortin-4 receptor agonist. The patients had a sustainable reduction in hunger and substantial weight loss (51.0 kg after 42 weeks in Patient 1 and 20.5 kg after 12 weeks in Patient 2).


Subject(s)
Hyperphagia/drug therapy , Metabolism, Inborn Errors/drug therapy , Obesity/drug therapy , Pro-Opiomelanocortin/deficiency , Receptor, Melanocortin, Type 4/agonists , alpha-MSH/analogs & derivatives , Adult , Blood Pressure , Female , Humans , Hyperphagia/genetics , Metabolism, Inborn Errors/genetics , Obesity/genetics , Phenotype , Pilot Projects , Pro-Opiomelanocortin/genetics , Young Adult , alpha-MSH/adverse effects , alpha-MSH/therapeutic use
20.
J Neuroinflammation ; 16(1): 192, 2019 Oct 28.
Article in English | MEDLINE | ID: mdl-31660977

ABSTRACT

BACKGROUND: Neuroinflammation and blood-brain barrier (BBB) disruption are two vital mechanisms of secondary brain injury following intracerebral hemorrhage (ICH). Recently, melanocortin-1 receptor (Mc1r) activation by Nle4-D-Phe7-α-MSH (NDP-MSH) was shown to play a neuroprotective role in an experimental autoimmune encephalomyelitis (EAE) mouse model. This study aimed to investigate whether NDP-MSH could alleviate neuroinflammation and BBB disruption after experimental ICH, as well as the potential mechanisms of its neuroprotective roles. METHODS: Two hundred and eighteen male C57BL/6 mice were subjected to autologous blood-injection ICH model. NDP-MSH, an agonist of Mc1r, was administered intraperitoneally injected at 1 h after ICH insult. To further explore the related protective mechanisms, Mc1r small interfering RNA (Mc1r siRNA) and nuclear receptor subfamily 4 group A member 1 (Nr4a1) siRNA were administered via intracerebroventricular (i.c.v) injection before ICH induction. Neurological test, BBB permeability, brain water content, immunofluorescence staining, and Western blot analysis were implemented. RESULTS: The Expression of Mc1r was significantly increased after ICH. Mc1r was mainly expressed in microglia, astrocytes, and endothelial cells following ICH. Treatment with NDP-MSH remarkably improved neurological function and reduced BBB disruption, brain water content, and the number of microglia in the peri-hematoma tissue after ICH. Meanwhile, the administration of NDP-MSH significantly reduced the expression of p-NF-κB p65, IL-1ß, TNF-α, and MMP-9 and increased the expression of p-CREB, Nr4a1, ZO-1, occludin, and Lama5. Inversely, the knockdown of Mc1r or Nr4a1 abolished the neuroprotective effects of NDP-MSH. CONCLUSIONS: Taken together, NDP-MSH binding Mc1r attenuated neuroinflammation and BBB disruption and improved neurological deficits, at least in part through CREB/Nr4a1/NF-κB pathway after ICH.


Subject(s)
Blood-Brain Barrier/metabolism , Cerebral Hemorrhage/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , NF-kappa B/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Receptor, Melanocortin, Type 1/metabolism , alpha-MSH/analogs & derivatives , Animals , Blood-Brain Barrier/drug effects , Brain/drug effects , Brain/metabolism , Male , Mice , Mice, Inbred C57BL , Protein Binding/drug effects , Protein Binding/physiology , Signal Transduction/drug effects , Signal Transduction/physiology , alpha-MSH/metabolism , alpha-MSH/pharmacology , alpha-MSH/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL