Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 73
Filtrer
1.
medRxiv ; 2024 Jun 14.
Article de Anglais | MEDLINE | ID: mdl-38946973

RÉSUMÉ

Individuals with Down syndrome (DS), the genetic condition caused by trisomy 21 (T21), display clear signs of immune dysregulation, including high rates of autoimmune disorders and severe complications from infections. Although it is well established that T21 causes increased interferon responses and JAK/STAT signaling, elevated autoantibodies, global immune remodeling, and hypercytokinemia, the interplay between these processes, the clinical manifestations of DS, and potential therapeutic interventions remain ill defined. Here, we report a comprehensive analysis of immune dysregulation at the clinical, cellular, and molecular level in hundreds of individuals with DS. We demonstrate multi-organ autoimmunity of pediatric onset concurrent with unexpected autoantibody-phenotype associations. Importantly, constitutive immune remodeling and hypercytokinemia occur from an early age prior to autoimmune diagnoses or autoantibody production. We then report the interim analysis of a Phase II clinical trial investigating the safety and efficacy of the JAK inhibitor tofacitinib through multiple clinical and molecular endpoints. Analysis of the first 10 participants to complete the 16-week study shows a good safety profile and no serious adverse events. Treatment reduced skin pathology in alopecia areata, psoriasis, and atopic dermatitis, while decreasing interferon scores, cytokine scores, and levels of pathogenic autoantibodies without overt immune suppression. Additional research is needed to define the effects of JAK inhibition on the broader developmental and clinical hallmarks of DS. ClinicalTrials.gov identifier: NCT04246372.

2.
J Invest Dermatol ; 2024 Jun 06.
Article de Anglais | MEDLINE | ID: mdl-38848986

RÉSUMÉ

A better understanding of human melanocyte (MC) and MC stem cell biology is essential for treating MC-related diseases. This study employed an inherited pigmentation disorder carrying the SASH1S519N variant in a Hispanic family to investigate SASH1 function in the MC lineage and the underlying mechanism for this disorder. We used a multidisciplinary approach, including clinical examinations, human cell assays, yeast 2-hybrid screening, and biochemical techniques. Results linked early hair graying to the SASH1S519N variant, a previously unrecognized clinical phenotype in hyperpigmentation disorders. In vitro, we identified SASH1 as a regulator in MC stem cell maintenance and discovered that TNKS2 is crucial for SASH1's role. In addition, the S519N variant is located in one of multiple tankyrase-binding motifs and alters the binding kinetics and affinity of the interaction. In summary, this disorder links both gain and loss of pigmentation in the same individual, hinting to accelerated aging in human MC stem cells. The findings offer insights into the roles of SASH1 and TNKS2 in MC stem cell maintenance and the molecular mechanisms of pigmentation disorders. We propose that a comprehensive clinical evaluation of patients with MC-related disorders should include an assessment and history of hair pigmentation loss.

3.
Cell Death Dis ; 15(3): 198, 2024 Mar 08.
Article de Anglais | MEDLINE | ID: mdl-38459020

RÉSUMÉ

Immune checkpoint inhibitors (ICIs) are now the first-line treatment for patients with advanced melanoma. Despite promising clinical results, many patients fail to respond to these therapies. BH3 mimetics, a novel class of small molecule inhibitors that bind and inhibit anti-apoptotic members of the BCL2 family proteins such as BCL2 or MCL1, have been very successful in treating hematologic malignancies. However, there are limited studies on the immunomodulatory role of the BH3 mimetics. Several factors contribute to ICI resistance including myeloid-derived suppressor cells (MDSCs) that exert immunosuppressive effects through direct and indirect inhibition of antitumor immunity. Thus, targeting MDSCs to enhance antitumor immunity has the potential to enhance the efficacy of ICIs. In this study, we show that the MCL1 inhibitor S64315 reduces melanoma tumor growth in an immune cell-dependent manner in mice. Specifically, S64315 enhances antitumor immunity by reducing MDSC frequency and by promoting the activity of CD8+T cells. Additionally, human MDSCs are 10 times more sensitive to S64315 than cutaneous melanoma lines. Further, we found that a higher expression of MCL1 is associated with poor survival for patients treated with anti-PD-1. Finally, combining S64315 and anti-PD-1 significantly slowed tumor growth compared to either agent alone. Together, this proof-of-concept study demonstrates the potential of combining an MCL1 inhibitor with anti-PD-1 in the treatment of melanoma. It justifies the further development of next generation MCL1 inhibitors to improve efficacy of ICIs in treating malignant melanoma.


Sujet(s)
Antinéoplasiques , Mélanome , Cellules myéloïdes suppressives , Tumeurs cutanées , Humains , Animaux , Souris , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Protéine Mcl-1/métabolisme , Mélanome/traitement médicamenteux , Antinéoplasiques/pharmacologie , Lymphocytes T CD8+/métabolisme , Cellules myéloïdes suppressives/métabolisme
4.
Pigment Cell Melanoma Res ; 37(3): 378-390, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38343115

RÉSUMÉ

We have discovered that human vitiligo patients treated with narrow-band UVB (NBUVB) demonstrated localized resistance to repigmentation in skin sites characterized by distinct cellular and molecular pathways. Using immunostaining studies, discovery-stage RNA-Seq analysis, and confirmatory in situ hybridization, we analyzed paired biopsies collected from vitiligo lesions that did not repigment after 6 months of NBUVB treatment (non-responding) and compared them with repigmented (responding) lesions from the same patient. Non-responding lesions exhibited acanthotic epidermis, had low number of total, proliferative, and differentiated melanocyte (MC) populations, and increased number of senescent keratinocytes (KCs) and of cytotoxic CD8+ T cells as compared with responding lesions. The abnormal response in the non-responding lesions was driven by a dysregulated cAMP pathway and of upstream activator PDE4B, and of WNT/ß-catenin repigmentation pathway. Vitiligo-responding lesions expressed high levels of WNT10B ligand, a molecule that may prevent epidermal senescence induced by NBUVB, and that in cultured melanoblasts prevented the pro-melanogenic effect of α-MSH. Understanding the pathways that govern lack of NBUVB-induced vitiligo repigmentation has a great promise in guiding the development of new therapeutic strategies for vitiligo.


Sujet(s)
Épiderme , Mélanocytes , Pigmentation de la peau , Vitiligo , Vitiligo/anatomopathologie , Vitiligo/radiothérapie , Vitiligo/métabolisme , Humains , Épiderme/anatomopathologie , Épiderme/métabolisme , Épiderme/effets des radiations , Pigmentation de la peau/effets des radiations , Mélanocytes/anatomopathologie , Mélanocytes/métabolisme , Mélanocytes/effets des radiations , Traitement par ultraviolets/méthodes , Kératinocytes/métabolisme , Kératinocytes/anatomopathologie , Kératinocytes/effets des radiations , Rayons ultraviolets , Femelle , Mâle , Voie de signalisation Wnt , Cyclic Nucleotide Phosphodiesterases, Type 4/métabolisme , Cyclic Nucleotide Phosphodiesterases, Type 4/génétique
5.
bioRxiv ; 2023 Sep 27.
Article de Anglais | MEDLINE | ID: mdl-37808724

RÉSUMÉ

Both aging spots (hyperpigmentation) and hair graying (lack of pigmentation) are associated with aging, two seemingly opposite pigmentation phenotypes. It is not clear how they are mechanistically connected. This study investigated the underlying mechanism in a family with an inherited pigmentation disorder. Clinical examinations identified accelerated hair graying and skin dyspigmentation (intermixed hyper and hypopigmentation) in the family members carrying the SASH1 S519N variant. Cell assays indicated that SASH1 promoted stem-like characteristics in human melanocytes, and SASH1 S519N was defective in this function. Multiple assays showed that SASH1 binds to tankyrase 2 (TNKS2), which is required for SASH1's promotion of stem-like function. Further, the SASH1 S519N variant is in a bona fide Tankyrase-binding motif, and SASH1 S519N alters the binding kinetics and affinity. Results here indicate SASH1 as a novel protein regulating the appropriate balance between melanocyte stem cells (McSC) and mature melanocytes (MCs), with S519N variant causing defects. We propose that dysfunction of McSC maintenance connects multiple aging-associated pigmentation phenotypes in the general population.

6.
J Invest Dermatol ; 142(7): 1912-1922.e7, 2022 07.
Article de Anglais | MEDLINE | ID: mdl-34942200

RÉSUMÉ

Uveal melanoma (UM) is a subtype of melanoma. Although they share a melanocytic origin with cutaneous melanoma (CM), patients with UM have few treatment options. BCL2 homologous 3 mimetics are small-molecule drugs that mimic proapoptotic BCL2 family members. We compared BCL2 family member expression between UM and CM using immunoblot and The Cancer Genome Atlas transcriptomic analysis. UM has a unique signature of low BFL1 and high PUMA proteins compared with CM and 30 other cancer types, making them an attractive candidate for BCL2 homologous 3 protein mimetics. We tested the efficacy of a BCL2 inhibitor and MCL1 inhibitor (MCL1i) in UM, with viability assays, live-cell imaging, sphere assays, and mouse xenograft models. UM had a higher sensitivity to MCL1i than CM. Overexpression of BFL1 or knockdown of PUMA made the UM more resistant to MCL1i. In contrast, MAPK/extracellular signal‒regulated kinase inhibitor treatment in CM made them more sensitive to MCL1i. However, MCL1i-alone treatment was not very effective to reduce the UM initiating cells; to overcome this, we employed a combination of MCL1i with BCL2 inhibitor that synergistically inhibited UM initiating cell's capacity to expand. Overall, we identify a distinct expression profile of BCL2 family members for UM that makes them susceptible to BCL2 homologous 3 mimetics.


Sujet(s)
Antinéoplasiques , Mélanome , Tumeurs cutanées , Animaux , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Humains , Mélanome/traitement médicamenteux , Mélanome/génétique , Souris , Protéines proto-oncogènes c-bcl-2 , Tumeurs cutanées/traitement médicamenteux , Tumeurs cutanées/génétique , Tumeurs de l'uvée ,
7.
Pharmaceuticals (Basel) ; 14(8)2021 Jul 30.
Article de Anglais | MEDLINE | ID: mdl-34451846

RÉSUMÉ

Although treatment options for melanoma patients have expanded in recent years with the approval of immunotherapy and targeted therapy, there is still an unmet need for new treatment options for patients that are ineligible for, or resistant to these therapies. BH3 mimetics, drugs that mimic the activity of pro-apoptotic BCL2 family proteins, have recently achieved remarkable success in the clinical setting. The combination of BH3 mimetic ABT-199 (venetoclax) plus azacitidine has shown substantial benefit in treating acute myelogenous leukemia. We evaluated the efficacy of various combinations of BH3 mimetic + azacitidine in fourteen human melanoma cell lines from cutaneous, mucosal, acral and uveal subtypes. Using a combination of cell viability assay, BCL2 family knockdown cell lines, live cell imaging, and sphere formation assay, we found that combining inhibition of MCL1, an anti-apoptotic BCL2 protein, with azacitidine had substantial pro-apoptotic effects in multiple melanoma cell lines. Specifically, this combination reduced cell viability, proliferation, sphere formation, and induced apoptosis. In addition, this combination is highly effective at reducing cell viability in rare mucosal and uveal subtypes. Overall, our data suggest this combination as a promising therapeutic option for some patients with melanoma and should be further explored in clinical trials.

8.
Am J Med Genet A ; 185(11): 3390-3400, 2021 11.
Article de Anglais | MEDLINE | ID: mdl-34435747

RÉSUMÉ

Recessive dystrophic epidermolysis bullosa (RDEB) is a rare genodermatosis caused by mutations in the gene coding for type VII collagen (COL7A1). More than 800 different pathogenic mutations in COL7A1 have been described to date; however, the ancestral origins of many of these mutations have not been precisely identified. In this study, 32 RDEB patient samples from the Southwestern United States, Mexico, Chile, and Colombia carrying common mutations in the COL7A1 gene were investigated to determine the origins of these mutations and the extent to which shared ancestry contributes to disease prevalence. The results demonstrate both shared European and American origins of RDEB mutations in distinct populations in the Americas and suggest the influence of Sephardic ancestry in at least some RDEB mutations of European origins. Knowledge of ancestry and relatedness among RDEB patient populations will be crucial for the development of future clinical trials and the advancement of novel therapeutics.


Sujet(s)
Collagène de type VII/génétique , Épidermolyse bulleuse dystrophique/génétique , Hispanique ou Latino/génétique , Juif/génétique , Chili/épidémiologie , Colombie/épidémiologie , Épidermolyse bulleuse dystrophique/épidémiologie , Femelle , Gènes récessifs/génétique , Humains , Mâle , Mexique/épidémiologie , Phénotype , États-Unis/épidémiologie
9.
Methods Mol Biol ; 2265: 185-199, 2021.
Article de Anglais | MEDLINE | ID: mdl-33704715

RÉSUMÉ

Sphere assays are widely used in vitro techniques to enrich and evaluate the stem-like cell behavior of both normal and cancer cells. Utilizing three-dimensional in vitro sphere culture conditions provide a better representation of tumor growth in vivo than the more common monolayer cultures. We describe how to perform primary and secondary sphere assays, used for the enrichment and self-renewability studies of melanoma/melanocyte stem-like cells. Spheres are generated by growing melanoma cells at low density in nonadherent conditions with stem cell media. We provide protocols for preparing inexpensive and versatile polyHEMA-coated plates, setting up primary and secondary sphere assays in almost any tissue culture format and quantification methods using standard inverted microscopy. Our protocol is easily adaptable to laboratories with basic cell culture capabilities, without the need for expensive fluidic instruments.


Sujet(s)
Techniques de culture cellulaire/méthodes , Mélanome/anatomopathologie , Cellules souches tumorales/cytologie , Sphéroïdes de cellules/anatomopathologie , Animaux , Lignée cellulaire tumorale , Survie cellulaire/effets des médicaments et des substances chimiques , Humains , Souris , Cellules souches tumorales/anatomopathologie , Tests d'activité antitumorale sur modèle de xénogreffe
10.
J Invest Dermatol ; 141(3): 638-647.e13, 2021 03.
Article de Anglais | MEDLINE | ID: mdl-32800877

RÉSUMÉ

In repigmentation of human vitiligo, the melanocyte (MC) precursors in the hair follicle bulge proliferate, migrate, and differentiate to repopulate the depigmented epidermis. Here, we present a comprehensive characterization of pathways and signals in the bulge that control the repigmentation process. Using biopsies from patients with vitiligo, we have selectively harvested, by laser capture microdissection, MC and keratinocyte precursors from the hair follicle bulge of untreated vitiligo skin and vitiligo skin treated with narrow-band UVB. The captured material was subjected to whole transcriptome RNA-sequencing. With this strategy, we found that repigmentation in the bulge MC precursors is driven by KCTD10, a signal with unknown roles in the skin, and CTNNB1 (encoding ß-catenin) and RHO guanosine triphosphatase [RHO GTPase, RHO], two signaling pathways previously shown to be involved in pigmentation biology. Knockdown studies in cultured human MCs of RHOJ, the upmost differentially expressed RHO family component, corroborated with our findings in patients with vitiligo, identified RHOJ involvement in UV response and melanization, and confirmed previously identified roles in melanocytic cell migration and apoptosis. A better understanding of mechanisms that govern repigmentation in MC precursors will enable the discovery of molecules that induce robust repigmentation phenotypes in vitiligo.


Sujet(s)
Cellules souches adultes/métabolisme , Mélanocytes/métabolisme , Pigmentation de la peau/effets des radiations , Traitement par ultraviolets , Vitiligo/thérapie , Adolescent , Adulte , Cellules souches adultes/effets des radiations , Sujet âgé , Enfant , Femelle , Follicule pileux/cytologie , Follicule pileux/métabolisme , Follicule pileux/anatomopathologie , Follicule pileux/effets des radiations , Humains , Kératinocytes/métabolisme , Kératinocytes/effets des radiations , Mâle , Mélanocytes/effets des radiations , Adulte d'âge moyen , Canaux potassiques voltage-dépendants/métabolisme , RNA-Seq , Transduction du signal/effets des radiations , Résultat thérapeutique , Vitiligo/anatomopathologie , Jeune adulte , bêta-Caténine/métabolisme , Protéines G rho/métabolisme
11.
J Investig Dermatol Symp Proc ; 20(1): S1-S5, 2020 11.
Article de Anglais | MEDLINE | ID: mdl-33099376

RÉSUMÉ

Alopecia areata (AA) is a common autoimmune skin disease resulting in the loss of hair on the scalp and elsewhere on the body that affects over 146 million people worldwide at some point in their lives. Founded in 1981, the National AA Foundation (NAAF) is a nonprofit organization that supports research to find a cure or acceptable treatment for AA, supports those with the disease, and educates the public about AA. NAAF conducts research summits every two years to review progress and create new directions in its funded and promoted research. This report from the seventh AA Research Summit, Forging the Future, held December 4-5, 2018 in New York City provides highlights of the research presented and future research priorities identified during targeted discussion sessions.


Sujet(s)
Pelade/thérapie , Hidrosadénite suppurée/thérapie , , Psoriasis/thérapie , Arthrite psoriasique/thérapie , Essais cliniques comme sujet , Congrès comme sujet , Détermination du point final , Humains , Satisfaction des patients
12.
Cancers (Basel) ; 12(8)2020 Aug 05.
Article de Anglais | MEDLINE | ID: mdl-32764384

RÉSUMÉ

There is an urgent need to develop treatments for patients with melanoma who are refractory to or ineligible for immune checkpoint blockade, including patients who lack BRAF-V600E/K mutations. This is often the case in patients diagnosed with rare melanoma subtypes such as mucosal and acral melanoma. Here, we analyzed data from the cutaneous melanoma The Cancer Genome Atlas Network (TCGA) transcriptomic and proteomic databases for differential expression of apoptosis molecules between melanomas with or without BRAF hotspot mutations. Our data indicated higher B-cell CLL/lymphoma 2 (BCL2) expression in melanoma without BRAF hotspot mutations, suggesting that BH3 mimetics, such as ABT-199 (venetoclax, a small molecule against BCL2), may be a potential therapeutic option for these patients. We explored the efficacy of combining two BH3 mimetics, ABT-199 and a myeloid cell leukemia sequence 1 (MCL1) inhibitor (S63845 or S64315/MIK665) in cutaneous, mucosal and acral melanomas, in vitro and in vivo. Our data indicate this combination induced cell death in a broad range of melanoma cell lines, including melanoma initiating cell populations, and was more potent in melanoma cells without BRAF-V600E/K mutations. Our knockdown/knockout experiments suggest that several pro-apoptotic BCL2 family members, BCL2-like 11 (apoptosis facilitator) (BIM), phorbol-12-myristate-13-acetate-induced protein 1 (NOXA) or BID, play a role in the combination-induced effects. Overall, our study supports the rationale for combining an MCL1 inhibitor with a BCL2 inhibitor as a therapeutic option in patients with advanced melanoma.

13.
Cell Death Dis ; 11(6): 443, 2020 06 08.
Article de Anglais | MEDLINE | ID: mdl-32513939

RÉSUMÉ

Current treatment for patients with metastatic melanoma include molecular-targeted therapies and immune checkpoint inhibitors. However, a subset of melanomas are difficult-to-treat. These melanomas include those without the genetic markers for targeted therapy, non-responsive to immunotherapy, and those who have relapsed or exhausted their therapeutic options. Therefore, it is necessary to understand and explore other biological processes that may provide new therapeutic approaches. One of most appealing is targeting the apoptotic/anti-apoptotic system that is effective against leukemia. We used genetic knockdown and pharmacologic approaches of BH3 mimetics to target anti-apoptotic BCL2 family members and identified MCL1 and BCLXL as crucial pro-survival members in melanoma. We then examined the effects of combining BH3 mimetics to target MCL1 and BCLXL in vitro and in vivo. These include clinical-trial-ready compounds such as ABT-263 (Navitoclax) and S63845/S64315 (MIK655). We used cell lines derived from patients with difficult-to-treat melanomas. In vitro, the combined inhibition of MCL1 and BCLXL resulted in significantly effective cell killing compared to single-agent treatment (p < 0.05) in multiple assays, including sphere assays. The combination-induced cell death was independent of BIM, and NOXA. Recapitulated in our mouse xenograft model, the combination inhibited tumor growth, reduced sphere-forming capacity (p < 0.01 and 0.05, respectively), and had tolerable toxicity (p > 0.40). Taken together, this study suggests that dual targeting of MCL1 and BCLXL should be considered as a treatment option for difficult-to-treat melanoma patients.


Sujet(s)
Dérivés de l'aniline/usage thérapeutique , Antinéoplasiques/usage thérapeutique , Mélanome/traitement médicamenteux , Protéine Mcl-1/antagonistes et inhibiteurs , Sulfonamides/usage thérapeutique , Dérivés de l'aniline/pharmacologie , Animaux , Antinéoplasiques/pharmacologie , Humains , Souris , Souris nude , Sulfonamides/pharmacologie
14.
J Invest Dermatol ; 140(1): 29-37, 2020 01.
Article de Anglais | MEDLINE | ID: mdl-31196751

RÉSUMÉ

Vitiligo and alopecia areata (AA) are common autoimmune conditions characterized by white spots on the skin (vitiligo) and bald spots on the scalp (AA), which significantly impact patients' lives by damaging their appearance and function. Melanocytes are the target of immune destruction in vitiligo and are hypothesized to be the site of immune attack in AA. This inflammatory process can be partially reversed by immunosuppressive drugs. Both conditions demonstrate regenerative components that are just now being identified. In this review, we focus on the regenerative medicine aspects of vitiligo and AA, using experimental data from human, mouse, and in vitro models, summarizing the key pathways involved in repopulation of the epidermis with melanocytes in vitiligo and in regrowth of hair follicles in AA. We also discuss treatments that may activate these pathways. Of the regenerative treatments, JAK inhibitors and bimatoprost stimulate repopulation of depleted cells in both diseases, intralesional injections of autologous concentrated platelet-rich plasma and minoxidil showed some benefit in AA, and phototherapy with narrowband UVB was shown to be effective especially in vitiligo. Finally, we discuss future treatments based on the mobilization of stem cells to regenerate anagen hair follicles in AA and intraepidermal melanocytes in vitiligo.


Sujet(s)
Pelade/thérapie , Bimatoprost/usage thérapeutique , Régénération tissulaire guidée/méthodes , Follicule pileux/physiologie , Inhibiteurs des Janus kinases/usage thérapeutique , Mélanocytes/physiologie , Vitiligo/thérapie , Animaux , Mouvement cellulaire , Auto-renouvellement cellulaire , Humains , Souris , Photothérapie , Médecine régénérative
15.
JCI Insight ; 3(19)2018 10 04.
Article de Anglais | MEDLINE | ID: mdl-30282836

RÉSUMÉ

Alopecia areata (AA) is an autoimmune disease in which cytotoxic T cells specifically target growing hair follicles. We used high-throughput TCR sequencing in the C3H/HeJ mouse model of AA and in human AA patients to gain insight into pathogenic T cell populations and their dynamics, which revealed clonal CD8+ T cell expansions in lesional skin. In the C3H/HeJ model, we observed interindividual sharing of TCRß chain protein sequences, which strongly supports a model of antigenic drive in AA. The overlap between the lesional TCR repertoire and a population of CD8+NKG2D+ T cells in skin-draining lymph nodes identified this subset as pathogenic effectors. In AA patients, treatment with the oral JAK inhibitor tofacitinib resulted in a decrease in clonally expanded CD8+ T cells in the scalp but also revealed that many expanded lesional T cell clones do not completely disappear from either skin or blood during treatment with tofacitinib, which may explain in part the relapse of disease after stopping treatment.


Sujet(s)
Pelade/immunologie , Maladies auto-immunes/immunologie , Lymphocytes T CD8+/immunologie , Récepteurs aux antigènes des cellules T/génétique , Adolescent , Adulte , Pelade/traitement médicamenteux , Animaux , Lymphocytes T CD8+/effets des médicaments et des substances chimiques , Lymphocytes T CD8+/métabolisme , Modèles animaux de maladie humaine , Femelle , Études de suivi , Follicule pileux/cytologie , Follicule pileux/immunologie , Séquençage nucléotidique à haut débit , Humains , Noeuds lymphatiques/cytologie , Mâle , Souris , Souris de lignée C3H , Adulte d'âge moyen , Sous-famille K des récepteurs de cellules NK de type lectine/métabolisme , Projets pilotes , Pipéridines/administration et posologie , Pyrimidines/administration et posologie , Pyrroles/administration et posologie , Cuir chevelu , Résultat thérapeutique , Jeune adulte
16.
Pigment Cell Melanoma Res ; 31(6): 728-735, 2018 11.
Article de Anglais | MEDLINE | ID: mdl-30281213

RÉSUMÉ

In this perspective, we identify emerging frontiers in clinical and basic research of melanocyte biology and its associated biomedical disciplines. We describe challenges and opportunities in clinical and basic research of normal and diseased melanocytes that impact current approaches to research in melanoma and the dermatological sciences. We focus on four themes: (1) clinical melanoma research, (2) basic melanoma research, (3) clinical dermatology, and (4) basic pigment cell research, with the goal of outlining current highlights, challenges, and frontiers associated with pigmentation and melanocyte biology. Significantly, this document encapsulates important advances in melanocyte and melanoma research including emerging frontiers in melanoma immunotherapy, medical and surgical oncology, dermatology, vitiligo, albinism, genomics and systems biology, epidemiology, pigment biophysics and chemistry, and evolution.


Sujet(s)
Recherche biomédicale , Mélanocytes/anatomopathologie , Mélanome/anatomopathologie , Animaux , Modèles animaux de maladie humaine , Résistance aux médicaments antinéoplasiques , Humains , Mélanome/épidémiologie , Mélanome/prévention et contrôle , Mélanome/thérapie , Pigmentation
17.
Cell Death Dis ; 9(9): 907, 2018 09 05.
Article de Anglais | MEDLINE | ID: mdl-30185782

RÉSUMÉ

Despite the recent advancement in treating melanoma, options are still limited for patients without BRAF mutations or in relapse from current treatments. BH3 mimetics against members of the BCL-2 family have gained excitement with the recent success in hematological malignancies. However, single drug BH3 mimetic therapy in melanoma has limited effectiveness due to escape by the anti-apoptotic protein MCL-1 and/or survival of melanoma-initiating cells (MICs). We tested the efficacy of the BH3 mimetic combination of A-1210477 (an MCL-1 inhibitor) and ABT-263 (a BCL-2/BCL-XL/BCL-W inhibitor) in killing melanoma, especially MICs. We also sought to better define Dynamin-Related Protein 1 (DRP-1)'s role in melanoma; DRP-1 is known to interact with members of the BCL-2 family and is a possible therapeutic target for melanoma treatment. We used multiple assays (cell viability, apoptosis, bright field, immunoblot, and sphere formation), as well as the CRISPR/Cas9 genome-editing techniques. For clinical relevance, we employed patient samples of different mutation status, including some relapsed from current treatments such as anti-PD-1 immunotherapy. We found the BH3 mimetic combination kill both the MICs and non-MICs (bulk of melanoma) in all cell lines and patient samples irrespective of the mutation status or relapsed state (p < 0.05). Unexpectedly, the major pro-apoptotic proteins, NOXA and BIM, are not necessary for the combination-induced cell death. Furthermore, the combination impedes the activation of DRP-1, and inhibition of DRP-1 further enhances apoptosis (p < 0.05). DRP-1 effects in melanoma differ from those seen in other cancer cells. These results provide new insights into BCL-2 family's regulation of the apoptotic pathway in melanoma, and suggest that inhibiting the major anti-apoptotic proteins is sufficient to induce cell death even without involvement from major pro-apoptotic proteins. Importantly, our study also indicates that DRP-1 inhibition is a promising adjuvant for BH3 mimetics in melanoma treatment.


Sujet(s)
Apoptose/physiologie , dGTPases/métabolisme , Mélanome/métabolisme , Mélanome/anatomopathologie , Protéines associées aux microtubules/métabolisme , Protéines mitochondriales/métabolisme , Fragments peptidiques/métabolisme , Protéines proto-oncogènes/métabolisme , Protéines régulatrices de l'apoptose/métabolisme , Lignée cellulaire tumorale , Dynamines , Humains , Protéines proto-oncogènes c-bcl-2/métabolisme
18.
Mol Oncol ; 12(8): 1358-1373, 2018 08.
Article de Anglais | MEDLINE | ID: mdl-29879296

RÉSUMÉ

The carboxyl-terminal binding proteins (CtBP) are transcriptional corepressors that regulate the expression of multiple epithelial-specific and pro-apoptotic genes. Overexpression of CtBP occurs in many human cancers where they promote the epithelial-to-mesenchymal transition, stem cell-like features, and cell survival, while knockdown of CtBP in tumor cells results in p53-independent apoptosis. CtBPs are recruited to their target genes by binding to a conserved PXDLS peptide motif present in multiple DNA-binding transcription factors. Disrupting the interaction between CtBP and its transcription factor partners may be a means of altering CtBP-mediated transcriptional repression and a potential approach for cancer therapies. However, small molecules targeting protein-protein interactions have traditionally been difficult to identify. In this study, we took advantage of the fact that CtBP binds to a conserved peptide motif to explore the feasibility of using peptides containing the PXDLS motif fused to cell-penetrating peptides (CPP) to inhibit CtBP function. We demonstrate that these peptides disrupt the ability of CtBP to interact with its protein partner, E1A, in an AlphaScreen assay. Moreover, these peptides can enter both lung carcinoma and melanoma cells, disrupt the interaction between CtBP and a transcription factor partner, and inhibit CtBP-mediated transcriptional repression. Finally, the constitutive expression of one such peptide, Pep1-E1A-WT, in a melanoma cell line reverses CtBP-mediated oncogenic phenotypes including proliferation, migration, and sphere formation and limits tumor growth in vivo. Together, our results suggest that CPP-fused PXDLS-containing peptides can potentially be developed into a research tool or therapeutic agent targeting CtBP-mediated transcriptional events in various biological pathways.


Sujet(s)
Alcohol oxidoreductases/antagonistes et inhibiteurs , Peptides de pénétration cellulaire/pharmacologie , Protéines de liaison à l'ADN/antagonistes et inhibiteurs , Peptides/pharmacologie , Activation de la transcription/effets des médicaments et des substances chimiques , Alcohol oxidoreductases/métabolisme , Séquence d'acides aminés , Animaux , Lignée cellulaire tumorale , Peptides de pénétration cellulaire/composition chimique , Peptides de pénétration cellulaire/génétique , Protéines de liaison à l'ADN/métabolisme , Humains , Souris , Peptides/composition chimique , Peptides/génétique , Cartes d'interactions protéiques/effets des médicaments et des substances chimiques , Protéines de fusion recombinantes/composition chimique , Protéines de fusion recombinantes/génétique , Protéines de fusion recombinantes/pharmacologie , Facteur de transcription Zeb1/métabolisme
19.
J Am Acad Dermatol ; 78(5): 927-934.e6, 2018 05.
Article de Anglais | MEDLINE | ID: mdl-29678380

RÉSUMÉ

BACKGROUND: Digital dermoscopic image analysis of pigmented skin lesions (PSLs) has become increasingly popular, despite its unclear clinical utility. Unbiased, high-powered studies investigating the efficacy of commercially available systems are limited. OBJECTIVE: To investigate the diagnostic performance of the FotoFinder Mole-Analyzer in assessing PSLs for cutaneous melanoma. METHODS: In this 15-year retrospective study, the histopathologies of 1076 biopsied PSLs among a total of 2500 imaged PSLs were collected. The biopsied PSLs were categorized as benign or malignant (cutaneous melanoma) based on histopathology. Analyzer scores (0-1.00) for these PSLs were obtained and grouped according to histopathology. RESULTS: At an optimized cutoff score of 0.50, a sensitivity of 56% and a specificity of 74% were achieved. The area under the receiver operating characteristics curve was 0.698, indicating poor accuracy as a diagnostic tool. LIMITATIONS: This study had a retrospective design and involved only a single institution. CONCLUSION: Our study reveals a low sensitivity of the scoring function of this digital dermoscopic image analyzer for detecting cutaneous melanomas. Physicians must apply keen clinical judgment when using such devices in the screening of suspicious PSLs.


Sujet(s)
Dermoscopie/instrumentation , Diagnostic assisté par ordinateur/méthodes , Mélanome/anatomopathologie , Naevus pigmentaire/anatomopathologie , Tumeurs cutanées/anatomopathologie , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Aire sous la courbe , Ponction-biopsie à l'aiguille , Études de cohortes , Dermoscopie/méthodes , Diagnostic différentiel , Femelle , Humains , Immunohistochimie , Mâle , Mélanome/diagnostic , Adulte d'âge moyen , Naevus pigmentaire/diagnostic , Courbe ROC , Études rétrospectives , Sensibilité et spécificité , Tumeurs cutanées/diagnostic , Analyse et exécution des tâches ,
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE