Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Pigment Cell Melanoma Res ; 37(3): 378-390, 2024 May.
Article in English | MEDLINE | ID: mdl-38343115

ABSTRACT

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.


Subject(s)
Epidermis , Melanocytes , Skin Pigmentation , Vitiligo , Vitiligo/pathology , Vitiligo/radiotherapy , Vitiligo/metabolism , Humans , Epidermis/pathology , Epidermis/metabolism , Epidermis/radiation effects , Skin Pigmentation/radiation effects , Melanocytes/pathology , Melanocytes/metabolism , Melanocytes/radiation effects , Ultraviolet Therapy/methods , Keratinocytes/metabolism , Keratinocytes/pathology , Keratinocytes/radiation effects , Ultraviolet Rays , Female , Male , Wnt Signaling Pathway , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/genetics
2.
bioRxiv ; 2023 Sep 27.
Article in English | MEDLINE | ID: mdl-37808724

ABSTRACT

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.

3.
J Invest Dermatol ; 141(3): 638-647.e13, 2021 03.
Article in English | MEDLINE | ID: mdl-32800877

ABSTRACT

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.


Subject(s)
Adult Stem Cells/metabolism , Melanocytes/metabolism , Skin Pigmentation/radiation effects , Ultraviolet Therapy , Vitiligo/therapy , Adolescent , Adult , Adult Stem Cells/radiation effects , Aged , Child , Female , Hair Follicle/cytology , Hair Follicle/metabolism , Hair Follicle/pathology , Hair Follicle/radiation effects , Humans , Keratinocytes/metabolism , Keratinocytes/radiation effects , Male , Melanocytes/radiation effects , Middle Aged , Potassium Channels, Voltage-Gated/metabolism , RNA-Seq , Signal Transduction/radiation effects , Treatment Outcome , Vitiligo/pathology , Young Adult , beta Catenin/metabolism , rho GTP-Binding Proteins/metabolism
4.
J Invest Dermatol ; 140(1): 29-37, 2020 01.
Article in English | MEDLINE | ID: mdl-31196751

ABSTRACT

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.


Subject(s)
Alopecia Areata/therapy , Bimatoprost/therapeutic use , Guided Tissue Regeneration/methods , Hair Follicle/physiology , Janus Kinase Inhibitors/therapeutic use , Melanocytes/physiology , Vitiligo/therapy , Animals , Cell Movement , Cell Self Renewal , Humans , Mice , Phototherapy , Regenerative Medicine
5.
J Invest Dermatol ; 138(3): 657-668, 2018 03.
Article in English | MEDLINE | ID: mdl-29054607

ABSTRACT

Vitiligo repigmentation is a complex process in which the melanocyte-depleted interfollicular epidermis is repopulated by melanocyte precursors from hair follicle bulge that proliferate, migrate, and differentiate into mature melanocytes on their way to the epidermis. The strongest stimulus for vitiligo repigmentation is narrow-band UVB (NBUVB), but how the hair follicle melanocyte precursors are activated by UV light has not been extensively studied. To better understand this process, we developed an application that combined laser capture microdissection and subsequent whole transcriptome RNA sequencing of hair follicle bulge melanocyte precursors and compared their gene signatures to that of regenerated mature epidermal melanocytes from NBUVB-treated vitiligo skin. Using this strategy, we found up-regulation of TNC, GJB6, and THBS1 in the hair follicle bulge melanocytes and of TYR in the epidermal melanocytes of the NBUVB-treated vitiligo skin. We validated these results by quantitative real-time-PCR using NBUVB-treated vitiligo skin and untreated normal skin. We also identified that GLI1, a candidate stem cell-associated gene, is significantly up-regulated in the melanocytes captured from NBUVB-treated vitiligo bulge compared with untreated vitiligo bulge. These signals are potential key players in the activation of bulge melanocyte precursors during vitiligo repigmentation.


Subject(s)
Hair Follicle/cytology , Signal Transduction/physiology , Skin Pigmentation , Stem Cells/metabolism , Ultraviolet Therapy , Vitiligo/radiotherapy , Zinc Finger Protein GLI1/genetics , beta Catenin/physiology , Humans , Laser Capture Microdissection , Sequence Analysis, RNA , Transcription, Genetic
6.
Dermatol Clin ; 35(2): 205-218, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28317529

ABSTRACT

Repigmentation in vitiligo is the process that replaces, in the epidermal basal layer of vitiligo skin, the mature melanocytes that have been killed by cytotoxic T cells specific for melanocyte antigens. It consists of mobilization of melanocyte precursors in the hair follicle bulge and infundibulum to proliferate, migrate, and differentiate into mature melanocytes, moving from the hair follicle bulge to the interfollicular epidermis. The most common clinical presentation of repigmentation in vitiligo is the perifollicular pattern. The most potent stimulus for repigmentation is the UV light.


Subject(s)
Melanocytes , Regeneration , Ultraviolet Therapy , Vitiligo/therapy , Epidermal Cells , Hair Follicle/cytology , Humans , Keratinocytes , PUVA Therapy
7.
Exp Dermatol ; 25(10): 805-11, 2016 10.
Article in English | MEDLINE | ID: mdl-27193292

ABSTRACT

To characterize the gene expression profile of regenerated melanocytes in the narrow band UVB (NBUVB)-treated vitiligo epidermis and their precursors in the hair follicle, we present here a strategy of RNA isolation from in situ melanocytes using human frozen skin. We developed a rapid immunostaining protocol using the NKI-beteb antibody, which labels differentiated and precursor melanocytes, followed by fluorescent laser capture microdissection. This technique enabled the direct isolation, from melanocyte and adjacent keratinocyte populations, of satisfactory quality RNA that was successfully amplified and analysed by qRT-PCR. The melanocyte-specific gene transcripts TYR, DCT, TYRP1 and PMEL were significantly upregulated in our NBUVB-treated melanocyte samples as compared with the keratinocyte samples, while keratinocyte-specific genes (KRT5 and KRT14) were expressed significantly higher in the keratinocyte samples as compared with the melanocyte samples. Furthermore, in both NBUVB-treated vitiligo skin and normal skin, when bulge melanocytes were compared with epidermal melanocytes, we found significantly lower expression of melanocyte-specific genes and significantly higher expression of three melanocytic stem cell genes (SOX9, WIF1 and SFRP1), while ALCAM and ALDH1A1 transcripts did not show significant variation. We found significantly higher expression of melanocyte-specific genes in the epidermis of NBUVB-treated vitiligo, as compared to the normal skin. When comparing bulge melanocyte samples from untreated vitiligo, NBUVB-treated vitiligo and normal skin, we did not find significant differences in the expression of melanocyte-specific genes or melanocytic stem cell genes. These techniques offer valuable opportunities to study melanocytes and their precursors in vitiligo and other pigmentation disorders.


Subject(s)
Laser Capture Microdissection , Melanocytes/metabolism , RNA/isolation & purification , Vitiligo/metabolism , Case-Control Studies , Humans , RNA/metabolism , Vitiligo/radiotherapy
8.
J Invest Dermatol ; 135(8): 2068-2076, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25822579

ABSTRACT

In vitiligo, the autoimmune destruction of epidermal melanocytes produces white spots that can be repigmented by melanocyte precursors from the hair follicles, following stimulation with UV light. We examined by immunofluorescence the distribution of melanocyte markers (C-KIT, DCT, PAX3, and TYR) coupled with markers of proliferation (KI-67) and migration (MCAM) in precursors and mature melanocytes from the hair follicle and the epidermis of untreated and narrow band UVB (NBUVB)-treated human vitiligo skin. NBUVB was associated with a significant increase in the number of melanocytes in the infundibulum and with restoration of the normal melanocyte population in the epidermis, which was lacking in the untreated vitiligo. We identified several precursor populations (melanocyte stem cells, melanoblasts, and other immature phenotypes), and progressively differentiating melanocytes, some with putative migratory and/or proliferative abilities. The primary melanocyte germ was present in the untreated and treated hair follicle bulge, whereas a possible secondary melanocyte germ composed of C-KIT+ melanocytes was found in the infundibulum and interfollicular epidermis of UV-treated vitiligo. This is an exceptional model for studying the mobilization of melanocyte stem cells in human skin. Improved understanding of this process is essential for designing better treatments for vitiligo, ultimately based on melanocyte stem cell activation and mobilization.


Subject(s)
Melanocytes/pathology , Stem Cells/pathology , Ultraviolet Rays , Ultraviolet Therapy , Vitiligo/pathology , Vitiligo/radiotherapy , Cell Differentiation/radiation effects , Cell Movement/radiation effects , Cell Proliferation/radiation effects , Epidermis/metabolism , Epidermis/pathology , Epidermis/radiation effects , Hair Follicle/metabolism , Hair Follicle/pathology , Hair Follicle/radiation effects , Humans , Intramolecular Oxidoreductases/metabolism , Melanocytes/metabolism , Melanocytes/radiation effects , PAX3 Transcription Factor , Paired Box Transcription Factors/metabolism , Phenotype , Proto-Oncogene Proteins c-kit/metabolism , Stem Cells/metabolism , Stem Cells/radiation effects , Vitiligo/metabolism
9.
J Clin Invest ; 123(10): 4390-404, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23999427

ABSTRACT

Squamous cell carcinomas (SCCs) originate in stratified epithelia, with a small subset becoming metastatic. Epithelial stem cells are targets for driver mutations that give rise to SCCs, but it is unknown whether they contribute to oncogenic multipotency and metastasis. We developed a mouse model of SCC by targeting two frequent genetic mutations in human SCCs, oncogene Kras(G12D) activation and Smad4 deletion, to mouse keratin 15-expressing (K15+) stem cells. We show that transgenic mice developed multilineage tumors, including metastatic SCCs. Among cancer stem cell-enriched (CSC-enriched) populations, those with increased side population (SP) cells correlated with epithelial-mesenchymal transition (EMT) and lung metastasis. We show that microRNA-9 (miR-9) contributed to SP expansion and metastasis, and miR-9 inhibition reduced the number of SP cells and metastasis. Increased miR-9 was detected in metastatic human primary SCCs and SCC metastases, and miR-9-transduced human SCC cells exhibited increased invasion. We identified α-catenin as a predominant miR-9 target. Increased miR-9 in human SCC metastases correlated with α-catenin loss but not E-cadherin loss. Our results demonstrate that stem cells with Kras(G12D) activation and Smad4 depletion can produce tumors that are multipotent and susceptible to EMT and metastasis. Additionally, tumor initiation and metastatic properties of CSCs can be uncoupled, with miR-9 regulating the expansion of metastatic CSCs.


Subject(s)
Carcinoma, Squamous Cell/secondary , Neoplastic Stem Cells/pathology , Proto-Oncogene Proteins/genetics , Skin Neoplasms/pathology , Smad4 Protein/genetics , ras Proteins/genetics , Animals , Carcinogenesis/metabolism , Carcinoma, Squamous Cell/genetics , Cell Dedifferentiation , Cell Proliferation , Epithelial-Mesenchymal Transition , Female , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Nude , Mice, Transgenic , MicroRNAs/genetics , Mutation, Missense , Neoplasm Transplantation , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/physiology , Proto-Oncogene Proteins p21(ras) , RNA Interference , Sequence Deletion , Side-Population Cells/metabolism , Side-Population Cells/pathology , Side-Population Cells/physiology , Skin Neoplasms/genetics , Tumor Cells, Cultured , alpha Catenin/genetics , alpha Catenin/metabolism
10.
J Invest Dermatol ; 133(5): 1286-93, 2013 May.
Article in English | MEDLINE | ID: mdl-23190898

ABSTRACT

Melanoma is an aggressive cancer that metastasizes rapidly and is refractory to conventional chemotherapies. Identifying microRNAs (miRNAs) that are responsible for this pathogenesis is therefore a promising means of developing new therapies. We identified miR-26a through microarray and quantitative reverse-transcription-PCR (qRT-PCR) experiments as an miRNA that is strongly downregulated in melanoma cell lines as compared with primary melanocytes. Treatment of cell lines with miR-26a mimic caused significant and rapid cell death compared with a negative control in most melanoma cell lines tested. In surveying targets of miR-26a, we found that protein levels of SMAD1 (mothers against decapentaplegic homolog 1) and BAG-4/SODD were strongly decreased in sensitive cells treated with miR-26a mimic as compared with the control. The luciferase reporter assays further demonstrated that miR-26a can repress gene expression through the binding site in the 3' untranslated region (3'UTR) of SODD (silencer of death domains). Knockdown of these proteins with small interfering RNA (siRNA) showed that SODD has an important role in protecting melanoma cells from apoptosis in most cell lines sensitive to miR-26a, whereas SMAD1 may have a minor role. Furthermore, transfecting cells with a miR-26a inhibitor increased SODD expression. Our findings indicate that miR-26a replacement is a potential therapeutic strategy for metastatic melanoma, and that SODD, in particular, is a potentially useful therapeutic target.


Subject(s)
Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Apoptosis/physiology , Down-Regulation/physiology , Melanoma/metabolism , MicroRNAs/metabolism , Skin Neoplasms/metabolism , Adaptor Proteins, Signal Transducing/drug effects , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Down-Regulation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/physiology , Humans , Melanocytes/drug effects , Melanocytes/metabolism , Melanocytes/pathology , Melanoma/pathology , MicroRNAs/genetics , MicroRNAs/pharmacology , Microarray Analysis , Skin Neoplasms/pathology , Smad1 Protein/drug effects , Smad1 Protein/metabolism , Transfection
11.
Biol Open ; 1(2): 92-100, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-23213401

ABSTRACT

The BH3 mimetic ABT-737 is a potent inhibitor of the anti-apoptotic proteins Bcl-2, Bcl-X(L), and Bcl-w. The Bcl-2 family modulates sensitivity to anticancer drugs in many cancers, including melanomas. In this study, we examined whether ABT-737 is effective in killing melanoma cells either alone or in combination with a proteasome inhibitor already in clinical use (Bortezomib) in vitro and in vivo, and further evaluated the mechanisms of action. Results showed that ABT-737 alone induced modest cytotoxicity in melanoma cells, but only at higher doses. Knock-down of the anti-apoptotic proteins Bcl-2, Bcl-X(L), or Mcl-1 with siRNAs demonstrated that Mcl-1 is the critical mediator of melanoma's resistance to ABT-737 treatment. However, ABT-737 displayed strong synergistic lethality when combined with Bortezomib. Immunoblot analyses demonstrated that Bortezomib increased expression of Noxa, a pro-apoptotic Bcl-2 member that antagonizes Mcl-1. Additionally, siRNA-mediated inhibition of Noxa expression protected melanoma cells from cytotoxicity induced by the combination treatment. These results demonstrate that Bortezomib synergizes with ABT-737 by neutralizing Mcl-1's function via increased levels of Noxa. In a xenograft mouse model, although drug doses were limited due to toxicity, ABT-737 or Bortezomib slowed melanoma tumor growth compared to the control, and the drug combination significantly decreased growth compared to either drug alone. These data imply that less toxic drugs fulfilling a function similar to Bortezomib to neutralize Mcl-1 are promising candidates for combination with ABT-737 for treating melanomas.

12.
PLoS One ; 6(8): e24294, 2011.
Article in English | MEDLINE | ID: mdl-21897876

ABSTRACT

Metastatic melanoma has poor prognosis and is refractory to most conventional chemotherapies. The alkylating agent temozolomide (TMZ) is commonly used in treating melanoma but has a disappointing response rate. Agents that can act cooperatively with TMZ and improve its efficacy are thus highly sought after. The BH3 mimetic ABT-737, which can induce apoptosis by targeting pro-survival Bcl-2 family members, has been found to enhance the efficacy of many conventional chemotherapeutic agents in multiple cancers. We found that combining TMZ and ABT-737 induced strong synergistic apoptosis in multiple human melanoma cell lines. When the drugs were used in combination in a mouse xenograft model, they drastically reduced tumor growth at concentrations where each individual drug had no significant effect. We found that TMZ treatment elevated p53 levels, and that the pro-apoptotic protein Noxa was elevated in TMZ/ABT-737 treated cells. Experiments with shRNA demonstrated that the synergistic effect of TMZ and ABT-737 was largely dependent on Noxa. Experiments with nutlin-3, a p53 inducer, demonstrated that p53 induction was sufficient for synergistic cell death with ABT-737 in a Noxa-dependent fashion. However, p53 was not necessary for TMZ/ABT-737 synergy as demonstrated by a p53-null line, indicating that TMZ and ABT-737 together induce Noxa in a p53-independent fashion. These results demonstrate that targeting anti-apoptotic Bcl-2 members is a promising method for treating metastatic melanoma, and that clinical trials with TMZ and Bcl-2 inhibitors are warranted.


Subject(s)
Alkylating Agents/pharmacology , Apoptosis/drug effects , Biomimetic Materials/pharmacology , Biphenyl Compounds/pharmacology , Dacarbazine/analogs & derivatives , Melanoma/pathology , Nitrophenols/pharmacology , Sulfonamides/pharmacology , Animals , Cell Line, Tumor , Dacarbazine/pharmacology , Drug Synergism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Piperazines/pharmacology , Protein Structure, Tertiary , Proto-Oncogene Proteins c-bcl-2/metabolism , Temozolomide , Time Factors , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays
13.
J Invest Dermatol ; 129(4): 964-71, 2009 Apr.
Article in English | MEDLINE | ID: mdl-18987671

ABSTRACT

The Bcl-2 family is important in modulating sensitivity to anticancer drugs in many cancers, including melanomas. The BH3 mimetic ABT-737 is a potent small molecule inhibitor of the anti-apoptotic proteins Bcl-2/Bcl-X(L)/Bcl-w. In this report, we examined whether ABT-737 is effective in killing melanoma cells in combination with the proteasome inhibitor MG-132, and further evaluated the mechanisms of action. Viability, morphological, and Annexin V apoptosis assays showed that ABT-737 alone exhibited little cytotoxicity, yet it displayed strong synergistic lethality when combined with MG-132. In addition, the detection of Bax/Bak activation indicated that the combination treatment synergistically induced mitochondria-mediated apoptosis. Furthermore, mechanistic analysis revealed that this combination treatment induced expression of the pro-apoptotic protein Noxa- and caspase-dependent degradation of the anti-apoptotic protein, Mcl-1. Finally, siRNA-mediated inhibition of Mcl-1 expression significantly increased sensitivity to ABT-737 in these cells, and knocking down Noxa expression protected the cells from cytotoxicity induced by the combination treatment. These findings demonstrate that ABT-737 combined with MG-132 synergistically induced Noxa-dependent mitochondrial-mediated apoptosis. In summary, this study indicates promising therapeutic potential of targeting anti-apoptotic Bcl-2 family members in treating melanoma, and it validates rational molecular approaches that target anti-apoptotic defenses when developing cancer treatments.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Biphenyl Compounds/pharmacology , Cysteine Proteinase Inhibitors/pharmacology , Leupeptins/pharmacology , Melanoma/drug therapy , Nitrophenols/pharmacology , Proteasome Inhibitors , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Sulfonamides/pharmacology , Caspases/physiology , Cell Line, Tumor , Drug Synergism , Humans , Melanoma/pathology , Mitochondria/physiology , Myeloid Cell Leukemia Sequence 1 Protein , Piperazines/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-bcl-2/physiology
14.
J Invest Dermatol ; 129(2): 432-7, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18668139

ABSTRACT

B-Raf and N-Ras proteins are often activated in melanoma, yet their roles in producing inherent survival signals are not fully understood. In this study, we investigated how N-RAS(Q61K) and B-RAF(V600E) contribute to melanoma's resistance to apoptosis induced by detachment from the extracellular matrix (anoikis). We found that expression of constitutively active N-RAS(Q61K) and B-RAF(V600E) downregulated the proapoptotic Bim protein in an immortalized melanocyte cell line. Bim is one of the main proapoptotic mediators of anoikis. Western blot analysis showed that detachment increased Bim expression in melanocytes, and Annexin V staining indicated that detachment induced cell death significantly in melanocytes. Blocking Bim expression by using RNAi vectors or by expressing N-RAS(Q61K) significantly inhibited anoikis in melanocytes. In summary, this report indicates that N-RAS(Q61K) and B-RAF(V600E) contribute to melanoma's resistance to apoptosis in part by downregulating Bim expression, suggesting that Bim is a possible treatment target for overriding melanoma's inherent defenses against cell death.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Genes, ras/physiology , Melanocytes/physiology , Melanoma/physiopathology , Membrane Proteins/genetics , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins/genetics , Skin Neoplasms/physiopathology , Anoikis/physiology , Bcl-2-Like Protein 11 , Cell Line, Transformed , Down-Regulation/physiology , Gene Expression Regulation, Neoplastic/physiology , Humans , Melanocytes/cytology , Melanoma/pathology , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins c-akt/genetics , RNA Interference , Skin Neoplasms/pathology , Transfection
15.
J Invest Dermatol ; 128(4): 949-56, 2008 Apr.
Article in English | MEDLINE | ID: mdl-17989736

ABSTRACT

Hyperthermia has been revived as a promising approach for cancer treatment. To understand the underlying mechanisms of hyperthermic killing of cancer cells, we examined the cytotoxic effects of hyperthermia on various skin cancer cell lines using cell viability, morphological analyses, and caspase activation assays. Hyperthermia induced cytotoxicity in a time- and temperature-dependent manner. At middle dose/time combinations, heat-induced apoptosis, whereas at higher doses, necrosis was the mechanism of cell death. To investigate the mechanisms of hyperthermia-induced apoptosis, we examined the activation of extrinsic (Caspase 8) and intrinsic (Caspase 9) apoptotic pathways. Hyperthermia did not activate Caspases 8 or 9, but did activate Caspase 3/7, suggesting a non-conventional apoptotic pathway. Last, analysis of Grp78 expression and Caspase 12 or 4 activation indicated that hyperthermia induced endoplasmic reticulum-mediated apoptosis. Thus, hyperthermia induced apoptosis in two types of skin cancer cells through endoplasmic reticulum-mediated apoptosis and not through the classical intrinsic or extrinsic apoptosis pathways. Hyperthermia may be a promising treatment for basal cell carcinoma and melanoma, bypassing the antiapoptotic defenses concentrated in the intrinsic and extrinsic apoptosis pathways. These results also raise the possibility that heat may be combined with other approaches for induction of apoptosis to achieve synergistic killing of skin cancers.


Subject(s)
Apoptosis , Endoplasmic Reticulum/metabolism , Hyperthermia, Induced , Melanoma/therapy , Skin Neoplasms/therapy , Caspases/metabolism , Cell Line, Tumor , Endoplasmic Reticulum Chaperone BiP , Heat-Shock Proteins/metabolism , Humans , Melanoma/metabolism , Molecular Chaperones/metabolism , Skin Neoplasms/metabolism
16.
BMC Biotechnol ; 5: 12, 2005 May 10.
Article in English | MEDLINE | ID: mdl-15885144

ABSTRACT

BACKGROUND: Analyzing apoptosis has been an integral component of many biological studies. However, currently available methods for quantifying apoptosis have various limitations including multiple, sometimes cell-damaging steps, the inability to quantify live, necrotic and apoptotic cells at the same time, and non-specific detection (i.e. "false positive"). To overcome the shortcomings of current methods that quantify apoptosis in vitro and to take advantage of the 96-well plate format, we present here a modified ethidium bromide and acridine orange (EB/AO) staining assay, which may be performed entirely in a 96-well plate. Our method combines the advantages of the 96-well format and the conventional EB/AO method for apoptotic quantification. RESULTS: We compared our method and the conventional EB/AO method for quantifying apoptosis of suspension cells (Jurkat) and adherent cells (A375) under normal growth and apoptosis-inducing conditions. We found that our new EB/AO method achieved quantification results comparable to those produced using the conventional EB/AO method for both suspension and adherent cells. CONCLUSION: By eliminating the detaching and washing steps, our method drastically reduces the time needed to perform the test, minimizes damage to adherent cells, and decreases the possibility of losing floating cells. Overall, our method is an improvement over the currently available techniques especially for adherent cells.


Subject(s)
Apoptosis , Biotechnology/methods , Acridine Orange/pharmacology , Annexin A5/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Camptothecin/pharmacology , Caspase 3 , Caspase 7 , Caspases/biosynthesis , Cell Adhesion , Cell Line, Tumor , Chromatin/chemistry , Enzyme Inhibitors/pharmacology , Ethidium/pharmacology , False Positive Reactions , Fluorescent Dyes/pharmacology , Humans , In Situ Nick-End Labeling , Jurkat Cells , Necrosis , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...