Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 815
Filter
1.
Sci Rep ; 12(1): 4031, 2022 03 07.
Article in English | MEDLINE | ID: mdl-35256687

ABSTRACT

Angiosarcoma is a rare, life-threatening soft tissue sarcoma with malignant endothelial cells that is mainly found in the skin. Multidisciplinary approaches are used to treat patients with unresectable metastasized lesions; considering the cellular origin of angiosarcoma, anti-angiogenic therapy has also been used recently. However, these treatments have limited efficacy, and the survival rate remains low. Thus, more effective treatments need to be developed. Nectin cell adhesion molecule 4 (NECTIN4) is highly expressed in malignant tumors and promotes tumor progression. Thus, NECTIN4 is expected to be a novel therapeutic target for cancer. However, the significance of NECTIN4 in angiosarcoma remains unknown. Using immunohistochemistry, we investigated NECTIN4 expression in 74 tissue samples from angiosarcoma patients, finding variable NECTIN4 expression. In addition, we investigated NECTIN4 expression and function in human angiosarcoma cell lines. NECTIN4 expression was higher in angiosarcoma cells than normal endothelial cells, and angiosarcoma cells were sensitive to monomethyl auristatin E, the cytotoxic part of a NECTIN4-targetting antibody-drug conjugate. NECTIN4 knockdown inhibited the proliferation and angiogenesis of angiosarcoma cells, and Src kinase signaling was shown to be involved in NECTIN4 function, at least in part. NECTIN4-targeted therapy has the potential to be a novel treatment strategy for angiosarcoma.


Subject(s)
Cell Adhesion Molecules , Hemangiosarcoma , Skin Neoplasms , src-Family Kinases , Cell Adhesion Molecules/immunology , Cell Adhesion Molecules/metabolism , Endothelial Cells/metabolism , Hemangiosarcoma/blood supply , Hemangiosarcoma/drug therapy , Hemangiosarcoma/metabolism , Humans , Immunoconjugates/pharmacology , Nectins , Neovascularization, Pathologic/drug therapy , Skin Neoplasms/blood supply , Skin Neoplasms/drug therapy , Skin Neoplasms/metabolism , src-Family Kinases/metabolism
2.
Microvasc Res ; 140: 104305, 2022 03.
Article in English | MEDLINE | ID: mdl-34958805

ABSTRACT

Angiogenesis is critical for solid tumor growth beyond its minimal size. Previously, we reported that Down Syndrome Candidate Region 1 isoform 1L (DSCR1-1L) was one of the most up-regulated genes in endothelial cells induced by VEGF and histamine, and regulated endothelial cell proliferation, migration and angiogenesis. However, it was not known whether DSCR1-1L played a role in tumor growth. In this study, we found that DSCR1-1L shRNAs significantly inhibited the growth of transplanted melanoma in mice and its associated tumoral angiogenesis. In the gain of function assay, overexpression of DSCR1-1L cDNA in mouse endothelium is sufficient to significantly increase the tumor initiation induced by carcinogen, the growth of xenografted tumor, and the tumor metastasis in our endothelially-expressed DSCR1-1L transgenic mice, in which angiogenesis was induced. It was the first time to find that DSCR1-1L was also expressed in various tumor cells. DSCR1-1L shRNAs inhibited, but overexpression of DSCR1-1L cDNA increased, the tumor cell proliferation and migration. Most recently, we reported that DSCR1-1L modulated angiogenesis by down-regulation of VE-cadherin expression. Here, we found that DSCR1-1L down-regulated the expression of E-cadherin. Hence, DSCR1-1L is an excellent therapeutic target for cancers by regulation of both the endothelial and tumor cells through down-regulating (V)E-cadherin. DSCR1-1L shRNAs have the potential to be developed for clinical application.


Subject(s)
Calcium-Binding Proteins/metabolism , DNA-Binding Proteins/metabolism , Endothelial Cells/metabolism , Melanoma/blood supply , Melanoma/metabolism , Muscle Proteins/metabolism , Neovascularization, Pathologic , Skin Neoplasms/blood supply , Skin Neoplasms/metabolism , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Cadherins/genetics , Cadherins/metabolism , Calcium-Binding Proteins/genetics , Cell Line, Tumor , Cell Movement , Cell Proliferation , DNA-Binding Proteins/genetics , Endothelial Cells/pathology , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Melanoma/genetics , Melanoma/pathology , Mice, Nude , Muscle Proteins/genetics , Neoplasm Invasiveness , Protein Isoforms , Signal Transduction , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Tumor Burden
3.
Cells ; 10(10)2021 10 14.
Article in English | MEDLINE | ID: mdl-34685720

ABSTRACT

Different types of cells, such as endothelial cells, tumor-associated fibroblasts, pericytes, and immune cells, release extracellular vesicles (EVs) in the tumor microenvironment. The components of EVs include proteins, DNA, RNA, and microRNA. One of the most important functions of EVs is the transfer of aforementioned bioactive molecules, which in cancer cells may affect tumor growth, progression, angiogenesis, and metastatic spread. Furthermore, EVs affect the presentation of antigens to immune cells via the transfer of nucleic acids, peptides, and proteins to recipient cells. Recent studies have also explored the potential application of EVs in cancer treatment. This review summarizes the mechanisms by which EVs regulate melanoma development, progression, and their potentials to be applied in therapy. We initially describe vesicle components; discuss their effects on proliferation, anti-melanoma immunity, and drug resistance; and finally focus on the effects of EV-derived microRNAs on melanoma pathobiology. This work aims to facilitate our understanding of the influence of EVs on melanoma biology and initiate ideas for the development of novel therapeutic strategies.


Subject(s)
Extracellular Vesicles/metabolism , Melanoma/metabolism , Skin Neoplasms/metabolism , Animals , Disease Progression , Drug Resistance, Neoplasm , Humans , Melanoma/blood supply , Melanoma/pathology , Melanoma/therapy , Models, Biological , Skin Neoplasms/blood supply , Skin Neoplasms/pathology , Skin Neoplasms/therapy , Tumor Microenvironment , Melanoma, Cutaneous Malignant
4.
Biomolecules ; 11(6)2021 06 17.
Article in English | MEDLINE | ID: mdl-34204372

ABSTRACT

Basal cell carcinoma (BCC) is the most common skin malignancy, which rarely metastasizes but has a great ability to infiltrate and invade the surrounding tissues. One of the molecular players involved in the metastatic process are matrix metalloproteinases (MMPs). MMPs are enzymes that can degrade various components of the extracellular matrix. In the skin, the expression of MMPs is increased in response to various stimuli, including ultraviolet (UV) radiation, one of the main factors involved in the development of BCC. By modulating various processes that are linked to tumor growth, such as invasion and angiogenesis, MMPs have been associated with UV-related carcinogenesis. The sources of MMPs are multiple, as they can be released by both neoplastic and tumor microenvironment cells. Inhibiting the action of MMPs could be a useful therapeutic option in BCC management. In this review that reunites the latest advances in this domain, we discuss the role of MMPs in the pathogenesis and evolution of BCC, as molecules involved in tumor aggressiveness and risk of recurrence, in order to offer a fresh and updated perspective on this field.


Subject(s)
Carcinoma, Basal Cell , Collagenases/metabolism , Neoplasm Proteins/metabolism , Neovascularization, Pathologic , Skin Neoplasms , Tumor Microenvironment/radiation effects , Ultraviolet Rays/adverse effects , Carcinoma, Basal Cell/blood supply , Carcinoma, Basal Cell/enzymology , Carcinoma, Basal Cell/pathology , Humans , Neoplasm Invasiveness , Neovascularization, Pathologic/enzymology , Neovascularization, Pathologic/pathology , Skin Neoplasms/blood supply , Skin Neoplasms/enzymology , Skin Neoplasms/pathology
5.
Int J Mol Sci ; 22(13)2021 Jun 23.
Article in English | MEDLINE | ID: mdl-34201751

ABSTRACT

Cylindromatosis (CYLD) is a deubiquitinase (DUB) enzyme that was initially characterized as a tumor suppressor of adnexal skin tumors in patients with CYLD syndrome. Later, it was also shown that the expression of functionally inactive mutated forms of CYLD promoted tumor development and progression of non-melanoma skin cancer (NMSC). However, the ability of wild-type CYLD to inhibit skin tumorigenesis in vivo in immunocompetent mice has not been proved. Herein, we generated transgenic mice that express the wild type form of CYLD under the control of the keratin 5 (K5) promoter (K5-CYLDwt mice) and analyzed the skin properties of these transgenic mice by WB and immunohistochemistry, studied the survival and proliferating characteristics of primary keratinocytes, and performed chemical skin carcinogenesis experiments. As a result, we found a reduced activation of the nuclear factor kappa B (NF-κB) pathway in the skin of K5-CYLDwt mice in response to tumor necrosis factor-α (TNF-α); accordingly, when subjected to insults, K5-CYLDwt keratinocytes are prone to apoptosis and are protected from excessive hyperproliferation. Skin carcinogenesis assays showed inhibition of tumor development in K5-CYLDwt mice. As a mechanism of this tumor suppressor activity, we found that a moderate increase in CYLD expression levels reduced NF-κB activation, which favored the differentiation of tumor epidermal cells and inhibited its proliferation; moreover, it decreased tumor angiogenesis and inflammation. Altogether, our results suggest that increased levels of CYLD may be useful for anti-skin cancer therapy.


Subject(s)
Carcinoma, Squamous Cell/pathology , Deubiquitinating Enzyme CYLD/genetics , Skin Neoplasms/pathology , Animals , Carcinoma, Squamous Cell/blood supply , Carcinoma, Squamous Cell/genetics , Cell Differentiation/genetics , Cell Proliferation/genetics , Cells, Cultured , Deubiquitinating Enzyme CYLD/metabolism , Genes, Tumor Suppressor , Immunocompetence , Keratinocytes/drug effects , Keratinocytes/pathology , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Transgenic , NF-kappa B/metabolism , Neovascularization, Pathologic/genetics , Phorbol Esters/toxicity , Skin Neoplasms/blood supply , Skin Neoplasms/genetics , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology
6.
Mol Carcinog ; 60(3): 172-178, 2021 03.
Article in English | MEDLINE | ID: mdl-33482042

ABSTRACT

Although beta 2 adrenergic receptors (ß2 ADR) are present in the keratinocytes, their role in cutaneous squamous cell tumorigenesis needs to be ascertained. For the first time, we report here that selective ß2 ADR antagonists by inhibiting ß2 ADR actions significantly retarded the progression of ultraviolet B (UVB) induced premalignant cutaneous squamous cell lesions. These antagonists acted by inhibiting vascular endothelial growth factor-A (VEGF) mediated angiogenesis to prevent UVB radiation-induced squamous cell carcinoma of the skin.


Subject(s)
Adrenergic beta-2 Receptor Antagonists/pharmacology , Neoplasms, Squamous Cell/drug therapy , Skin Neoplasms/drug therapy , Ultraviolet Rays/adverse effects , Adrenergic beta-1 Receptor Agonists/pharmacology , Animals , Butoxamine/pharmacology , Humans , Keratinocytes/metabolism , Keratinocytes/radiation effects , Male , Mice, Inbred Strains , Neoplasms, Radiation-Induced/blood supply , Neoplasms, Radiation-Induced/drug therapy , Neoplasms, Radiation-Induced/etiology , Neoplasms, Squamous Cell/blood supply , Neoplasms, Squamous Cell/etiology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Receptors, Adrenergic, beta-2/metabolism , Skin Neoplasms/blood supply , Skin Neoplasms/etiology , Vascular Endothelial Growth Factor A/metabolism , Xamoterol/pharmacology
8.
J Invest Dermatol ; 141(4): 840-851.e4, 2021 04.
Article in English | MEDLINE | ID: mdl-32890629

ABSTRACT

Acquired BRAF/MAPK/extracellular signal‒regulated kinase inhibitor resistance in melanoma results in a new transcriptional state associated with an increased risk of metastasis. In this study, we identified noncanonical ephrin receptor (Eph) EphA2 signaling as a driver of the resistance-associated metastatic state. We used mass spectrometry‒based proteomic and phenotypic assays to demonstrate that the expression of active noncanonical EphA2-S897E in melanoma cells led to a mesenchymal-to-amoeboid transition driven by Cdc42 activation. The induction of mesenchymal-to-amoeboid transition promoted melanoma cell invasion, survival under shear stress, adhesion to endothelial cells under continuous-flow conditions, increased permeability of endothelial cell monolayers, and stimulated melanoma transendothelial cell migration. In vivo, melanoma cells expressing EphA2-S897E or active Cdc42 showed superior lung retention after tail-vain injection. Analysis of BRAF inhibitor‒sensitive and ‒resistant melanoma cells demonstrated resistance to be associated with a mesenchymal-to-amoeboid transition switch, upregulation of Cdc42 activity, increased invasion, and transendothelial migration. The drug-resistant metastatic state was dependent on histone deacetylase 8 activity. Silencing of histone deacetylase 8 led to the inhibition of EphA2 and protein kinase B phosphorylation, reduced invasion, and impaired melanoma cell-endothelial cell interactions. In summary, we have demonstrated that the metastatic state associated with acquired BRAF inhibitor resistance is dependent on noncanonical EphA2 signaling, leading to increased melanoma-endothelial cell interactions and enhanced tumor dissemination.


Subject(s)
Drug Resistance, Neoplasm/genetics , Melanoma/drug therapy , Protein Kinase Inhibitors/pharmacology , Receptor, EphA2/metabolism , Skin Neoplasms/drug therapy , Animals , Cell Communication/genetics , Cell Line, Tumor , Endothelial Cells/pathology , Endothelium, Vascular/cytology , Endothelium, Vascular/pathology , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Humans , Melanoma/blood supply , Melanoma/genetics , Melanoma/pathology , Mice , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/genetics , Receptor, EphA2/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction/genetics , Skin/blood supply , Skin/pathology , Skin Neoplasms/blood supply , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Transendothelial and Transepithelial Migration/genetics , Tumor Microenvironment/genetics , Xenograft Model Antitumor Assays
9.
J Cutan Pathol ; 48(2): 229-236, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32779250

ABSTRACT

In the last 30 years, there has been a strong interest in vascular proliferations. Pyogenic granuloma was not only renamed lobular capillary hemangioma, but also the conceptual interpretation was also changed from an overgrowth of granulation tissue to a genuine hemangioma (or benign vascular neoplasm). We describe 19 cases of patients who presented clinically with a vascular lesion, characteristically a pyogenic granuloma or lobular hemangioma, where the histopathological findings led to the pathologic concern for a lymphoma of the skin. These benign lesions with a dense lymphoid infiltrate were further defined on the basis of different vascular and lymphoid immunohistochemical markers as inflammatory lobular hemangiomas. We propose that given the considerable histopathological overlap between acral pseudolymphomatous angiokeratoma, T-cell rich angiomatoid polypoid pseudolymphoma of the skin, and other designations of some of these vascular proliferations with a rich and dense lymphoid infiltrate, they might constitute a spectrum of vascular lesions with varying clinical presentations.


Subject(s)
Cell Proliferation , Granuloma, Pyogenic , Hemangioma , Skin Neoplasms , T-Lymphocytes , Adolescent , Adult , Aged , Child , Female , Granuloma, Pyogenic/metabolism , Granuloma, Pyogenic/pathology , Hemangioma/blood supply , Hemangioma/metabolism , Hemangioma/pathology , Humans , Male , Middle Aged , Skin Neoplasms/blood supply , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology
10.
Adv Exp Med Biol ; 1287: 123-154, 2021.
Article in English | MEDLINE | ID: mdl-33034030

ABSTRACT

Since many decades, nonmelanoma skin cancer (NMSCs) is the most common malignancy worldwide. Basal cell carcinomas (BCC) and squamous cell carcinomas (SCC) are the major types of NMSCs, representing approximately 70% and 25% of these neoplasias, respectively. Because of their continuously rising incidence rates, NMSCs represent a constantly increasing global challenge for healthcare, although they are in most cases nonlethal and curable (e.g., by surgery). While at present, carcinogenesis of NMSC is still not fully understood, the relevance of genetic and molecular alterations in several pathways, including evolutionary highly conserved Notch signaling, has now been shown convincingly. The Notch pathway, which was first developed during evolution in metazoans and that was first discovered in fruit flies (Drosophila melanogaster), governs cell fate decisions and many other fundamental processes that are of high relevance not only for embryonic development, but also for initiation, promotion, and progression of cancer. Choosing NMSC as a model, we give in this review a brief overview on the interaction of Notch signaling with important oncogenic and tumor suppressor pathways and on its role for several hallmarks of carcinogenesis and cancer progression, including the regulation of cancer stem cells, tumor angiogenesis, and senescence.


Subject(s)
Carcinogenesis , Neoplastic Stem Cells/pathology , Neovascularization, Pathologic , Receptors, Notch/metabolism , Signal Transduction , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Animals , Carcinoma, Basal Cell/metabolism , Carcinoma, Basal Cell/pathology , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Humans , Skin Neoplasms/blood supply
11.
Exp Dermatol ; 29(11): 1046-1054, 2020 11.
Article in English | MEDLINE | ID: mdl-32998178

ABSTRACT

Metastatic melanoma is a devastating disease with a marginal-albeit increasing-hope for cure. Melanoma has a high mutation rate which correlates to the expression of numerous neo-antigens and thus is associated with the potential to induce and strengthen effective antitumoral immunity. However, the incomplete and potentially insufficient response to established immunotherapies (response rates usually do not markedly exceed 60%) already points to the need of further studies to improve treatment strategies. Multiple tumor escape mechanisms that allow melanoma to evade from antitumoral immune responses have been characterized and must be overcome to achieve a better clinical efficacy of immunotherapies. Recently, promising progress has been made in targeting tumor vasculature to control and increase the infiltration of tumors with effector lymphocytes. It has been hypothesized that amplified lymphocytic infiltrates in melanoma metastases result in a switch of the tumor microenvironment from a non-inflammatory to an inflammatory state. In this view point essay, we discuss the requirements for successful homing of lymphocytes to melanoma tissue and we present a mouse melanoma xenograft model that allows the investigation of human tumor vessels in vivo. Furthermore, current clinical studies dealing with the activation of melanoma vasculature for enhanced effectiveness of immunotherapy protocols are presented and open questions for routine clinical application are addressed.


Subject(s)
Endothelium/immunology , Immunotherapy , Lymphocytes/immunology , Melanoma/therapy , Skin Neoplasms/therapy , Animals , Blood Vessels/immunology , Cell Movement , Disease Models, Animal , Humans , Melanoma/blood supply , Melanoma/immunology , Melanoma/secondary , Mice , Receptors, Lymphocyte Homing , Skin Neoplasms/blood supply , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Tumor Escape/immunology
12.
Ann Diagn Pathol ; 48: 151597, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32829071

ABSTRACT

Angiosarcomas are ubiquitous neoplasms involving both cutaneous and soft tissue and visceral locations. Accumulating biomolecular evidences suggest that cutaneous angiosarcomas are distinctive entities with molecular, clinical and pathological peculiarities. Despite several ongoing clinical trials with promising therapeutic agents, the prognosis of cutaneous angiosarcomas is dismal and survival still rely on early diagnosis and surgery. An accurate diagnosis and the knowledge of the underlying molecular landscape are therefore essential to improve the prognosis. We detail the molecular, clinical, dermoscopic, morphological and prognostic features of cutaneous angiosarcoma. Although the molecular landscape of cutaneous angiosarcoma is not completely understood, accumulating evidences suggest that there are characteristic molecular alterations including dysregulation of angiogenesis and several complex molecular pathways. Secondary cutaneous angiosarcomas, arising in correlation with chronic lymphedema and ionizing radiation, have different molecular hallmarks, which are also leading to the first diagnostic applications. The diagnosis of cutaneous angiosarcoma may be challenging, as well-differentiated forms can be hard to distinguish from benign and low-grade vascular neoplasms, while poorly differentiated forms can be easily confounded with other non-vascular high-grade neoplasms. An accurate and early diagnosis, which is mandatory to ensure the best survival for the patients, is mainly based on morphological hallmarks.


Subject(s)
Early Detection of Cancer/standards , Hemangiosarcoma/diagnosis , Skin Neoplasms/blood supply , Skin Neoplasms/pathology , Aged , Chronic Disease , Dermoscopy/methods , Early Detection of Cancer/statistics & numerical data , Female , Hemangiosarcoma/etiology , Hemangiosarcoma/metabolism , Hemangiosarcoma/pathology , Humans , Lymphedema/complications , Middle Aged , Neoplasms, Radiation-Induced/pathology , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Pathology, Clinical/methods , Prognosis , Progression-Free Survival , Radiation, Ionizing , Skin Neoplasms/mortality
13.
Sci Rep ; 10(1): 11372, 2020 07 09.
Article in English | MEDLINE | ID: mdl-32647211

ABSTRACT

Administration of hemoglobin-based oxygen carriers (HBOCs) into the systemic circulation is a potential strategy to relieve solid tumor hypoxia in order to increase the effectiveness of chemotherapeutics. Previous computational analysis indicated that the oxygen (O2) status of the tumor and HBOC O2 affinity may play a role in increased O2 delivery to the tumor. However, no study has experimentally investigated how low- and high-affinity HBOCs would perform in normoxic and hypoxic tumors. In this study, we examined how the HBOC, polymerized human hemoglobin (PolyhHb), in the relaxed (R) or tense (T) quaternary state modulates O2 delivery to hypoxic (FME) and normoxic (LOX) human melanoma xenografts in a murine window chamber model. We examined microcirculatory fluid flow via video shearing optical microscopy, and O2 distributions via phosphorescence quenching microscopy. Additionally, we examined how weekly infusion of a 20% top-load dose of PolyhHb influences growth rate, vascularization, and regional blood flow in the FME and LOX tumor xenografts. Infusion of low-affinity T-state PolyhHb led to increased tissue oxygenation, decreased blood flow, decreased tumor growth, and decreased vascularization in hypoxic tumors. However, infusion of both T-state and R-state PolyhHbs led to worse outcomes in normoxic tumors. Of particular concern was the high-affinity R-state PolyhHb, which led to no improvement in hypoxic tumors and significantly worsened outcomes in normoxic tumors. Taken together, the results of this study indicate that the tumor O2 status is a primary determinant of the potency and outcomes of infused PolyhHb.


Subject(s)
Hemoglobins/pharmacology , Melanoma/drug therapy , Oxygen/metabolism , Polymers/pharmacology , Skin Neoplasms/drug therapy , Animals , Cell Hypoxia/drug effects , Erythrocytes/chemistry , Female , Hemoglobins/chemistry , Hemoglobins/isolation & purification , Hemoglobins/therapeutic use , Humans , Infusions, Intravenous , Melanoma/blood supply , Melanoma/pathology , Mice , Microcirculation/drug effects , Molecular Weight , Oxygen/analysis , Polymerization , Polymers/chemistry , Polymers/therapeutic use , Skin Neoplasms/blood supply , Skin Neoplasms/pathology , Tumor Microenvironment/drug effects , Xenograft Model Antitumor Assays
15.
J Ultrasound ; 23(1): 45-53, 2020 Mar.
Article in English | MEDLINE | ID: mdl-30927249

ABSTRACT

PURPOSE: Gel pad is an aqueous, flexible, easy available, disposable spacer used for the ultrasound (US) scan of superficial or difficult-to-visualize areas. In clinical practice, it is widely used in B-mode US approach of superficial lesions but, to date, no data have been provided as to its efficacy in the Doppler detection of superficial flows. The aim of our study was to demonstrate the role of stand-off gel pad in the detection of the otherwise-missed peri- or intra-lesional flow signals on Doppler imaging. MATERIALS AND METHODS: A total of 100 superficial lesions undergone to an US evaluation using a 7.5-12-MHz linear probe were evaluated prospectively with and without interposition of a gel stand-off pad to detect the presence or absence of vascularization and to classify the vascular pattern. RESULTS: Peri- or intra-lesional flow was demonstrated in 56% of cases without and in 84% of cases with interposition of a gel stand-off pad; moreover, a statistically significant difference (p value < 0.001) was observed at Chi-square test in the identification of the flow pattern between the use and no use of the pad. CONCLUSIONS: The use of a gel stand-off pad allows the detection of otherwise-missed peri- or intra-lesional flow signals on Doppler imaging, increasing the diagnostic role of this technique in differential diagnosis of superficial lesions.


Subject(s)
Skin Diseases/diagnostic imaging , Skin/diagnostic imaging , Ultrasonography, Doppler, Color/instrumentation , Adolescent , Adult , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Prospective Studies , Regional Blood Flow , Skin/blood supply , Skin/pathology , Skin Neoplasms/blood supply , Skin Neoplasms/diagnostic imaging , Ultrasonography, Doppler, Color/methods , Young Adult
16.
J Invest Dermatol ; 140(2): 445-454, 2020 02.
Article in English | MEDLINE | ID: mdl-31425704

ABSTRACT

Calpains, intracellular proteases specifically inhibited by calpastatin, play a major role in neoangiogenesis involved in tumor invasiveness and metastasis. They are partly exteriorized via the ATP-binding cassette transporter A1(ABCA1) transporter, but the importance of this process in tumor growth is still unknown. The aim of our study was to investigate the role of extracellular calpains in a model of melanoma by blocking their extracellular activity or exteriorization. In the first approach, a B16-F10 model of melanoma was developed in transgenic mice expressing high extracellular levels of calpastatin. In these mice, tumor growth was inhibited by ∼ 3-fold compared with wild-type animals. In vitro cytotoxicity assays and in vivo tumor studies have demonstrated that this protection was associated with a defect in tumor neoangiogenesis. Similarly, in wild-type animals given probenecid to blunt ABCA1 activity, melanoma tumor growth was inhibited by ∼ 3-fold. Again, this response was associated with a defect in neoangiogenesis. In vitro studies confirmed that probenecid limited endothelial cell migration and capillary formation from vascular explants. The observed reduction in fibronectin cleavage under these conditions is potentially involved in the response. Collectively, these studies demonstrate that probenecid, by blunting ABCA1 activity and thereby calpain exteriorization, limits melanoma tumor neoangiogenesis and invasiveness.


Subject(s)
ATP Binding Cassette Transporter 1/antagonists & inhibitors , Calpain/metabolism , Melanoma, Experimental/drug therapy , Neovascularization, Pathologic/drug therapy , Probenecid/pharmacology , Skin Neoplasms/drug therapy , ATP Binding Cassette Transporter 1/genetics , ATP Binding Cassette Transporter 1/metabolism , Animals , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cell Line, Tumor/transplantation , Cell Proliferation/drug effects , Humans , Male , Melanoma, Experimental/blood supply , Melanoma, Experimental/pathology , Mice , Mice, Transgenic , Neovascularization, Pathologic/pathology , Probenecid/therapeutic use , Skin Neoplasms/blood supply , Skin Neoplasms/pathology
17.
Pol J Pathol ; 70(3): 217-222, 2019.
Article in English | MEDLINE | ID: mdl-31820866

ABSTRACT

The study was aimed to evaluate the number of TAMs and to investigate whether they have association with microvessels density and patients' survival times. 46 cases of melanomas, divided into four groups according to the Breslow scale, were tested immunohistochemically with antibodies anti-CD68, CD163, iNOS to vizualized macrophages and anti-CD34 antibody to stain microvessels. The number of macrophages and the microvessels density were counted by hotspot analysis using an image analysis system. The study revealed increased numbers of CD68 and CD163 positive macrophages in successive stages of Breslow scale, but statistically significant differences were observed only between I and IV group for CD68 positive macrophages, and between I and III, IV group for CD163 positive macrophages. The mean number of the microvessels was significantly increased in group II, III, IV compared to group I. The correlative study showed significant positive correlations between the mean number of CD68 and CD163 positive macrophages and microvessels density. Moreover, the number of CD163 positive macrophages was associated inversely with patient's survival time. The results of our study may indicate that higher infiltration of macrophages, especially CD163 positive cells, is associated with more advanced melanomas, microvessels density and worse patient's prognosis.


Subject(s)
Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Macrophages/cytology , Melanoma/pathology , Receptors, Cell Surface/metabolism , Skin Neoplasms/pathology , Humans , Macrophages/metabolism , Melanoma/blood supply , Microvessels , Prognosis , Skin Neoplasms/blood supply
18.
Sci Rep ; 9(1): 18670, 2019 12 10.
Article in English | MEDLINE | ID: mdl-31822752

ABSTRACT

Microvascular networks of human basal cell carcinomas (BCC) and surrounding skin were assessed with optical coherence angiography (OCA) in conjunction with photodynamic therapy (PDT). OCA images were collected and analyzed in 31 lesions pre-treatment, and immediately/24 hours/3-12 months post-treatment. Pre-treatment OCA enabled differentiation between prevalent subtypes of BCC (nodular and superficial) and nodular-with-necrotic-core BCC subtypes with a diagnostic accuracy of 78%; this can facilitate more accurate biopsy reducing sampling error and better therapy regimen selection. Post-treatment OCA images at 24 hours were 98% predictive of eventual outcome. Additional findings highlight the importance of pre-treatment necrotic core, vascular metrics associated with hypertrophic scar formation, and early microvascular changes necessary in both tumorous and peri-tumorous regions to ensure treatment success.


Subject(s)
Angiography , Carcinoma, Basal Cell/diagnostic imaging , Carcinoma, Basal Cell/drug therapy , Photochemotherapy , Skin Neoplasms/diagnostic imaging , Skin Neoplasms/drug therapy , Tomography, Optical Coherence , Aged , Aged, 80 and over , Aminolevulinic Acid/administration & dosage , Carcinoma, Basal Cell/blood supply , Cohort Studies , Face/blood supply , Face/diagnostic imaging , Female , Humans , Male , Middle Aged , Necrosis , Photosensitizing Agents/administration & dosage , Skin/pathology , Skin Neoplasms/blood supply , Treatment Outcome
19.
Eur J Pharmacol ; 864: 172719, 2019 Dec 01.
Article in English | MEDLINE | ID: mdl-31586634

ABSTRACT

Gambogic acid (GA) is a potential anti-cancer compound that is extracted from the resin of Garciania hanburyi. The present study was designed to evaluate the anti-metastatic effect of GA on melanoma cell lines in vitro and to explore the underlying mechanism. The anti-proliferative activity of GA on melanoma cells was assessed by CCK-8 assay. The Wound-healing, transwell, adhesion, and tube formation assays were performed to examine the inhibition of GA on the cell's migration, invasion, adhesion, and angiogenesis capacities, respectively. Enzymatic activity of MMP-2 and MMP-9 were detected by gelatin zymography assay. Protein expressions regulated by GA treatment were tested by Western blot assay. The present results showed that GA significantly inhibited the proliferation of highly metastatic melanoma A375, B16-F10 cells and human umbilical vein endothelial cells (HUVECs) in time- and doses-dependent manners. Furthermore, GA significantly inhibited the migratory, invasive and adhesive properties of A375 and B16-F10 cells, and tube-forming potential of HUVECs at sub-IC50 concentrations, where no significant cytotoxicity was observed. Mechanistically, GA treatment suppressed the EMT and angiogenesis processes and reduced the enzymatic activity of MMP-2 and MMP-9. Moreover, abnormal PI3K/Akt and ERK signaling pathways in A375 and B16-F10 cells and HUVECs were notably suppressed by GA treatment. Collectively, our results suggest that GA exerts anti-metastasis activity in melanoma cells by suppressing the EMT and angiogenesis through the PI3K/Akt and ERK signaling pathways, and might be used as a phytomedicine against metastatic melanoma.


Subject(s)
Antineoplastic Agents/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Melanoma/pathology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Skin Neoplasms/pathology , Xanthones/pharmacology , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Humans , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Melanoma/blood supply , Mice , Neoplasm Invasiveness , Neoplasm Metastasis , Neovascularization, Pathologic/drug therapy , Skin Neoplasms/blood supply , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Xanthones/therapeutic use , Melanoma, Cutaneous Malignant
20.
J Dermatol Sci ; 96(1): 18-25, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31447183

ABSTRACT

BACKGROUND: Previous studies have indicated that MFG-E8 enhances tumor cell survival, invasion and angiogenesis. However, the role of MFG-E8 in angiosarcoma (AS) has not been clarified. OBJECTIVE: Objective was to elucidate the mechanism of the regulation by MFG-E8 in AS and the association between MFG-E8 and clinicopathological features of AS. METHODS: The effects of the depletion of MFG-E8 by siRNA on tube formation, migration and proliferation in murine AS cells were examined. The effect of administration of anti-MFG-E8 antibody (Ab) on tumor growth of AS in mice was examined. The associations of MFG-E8 expression and clinicopathological features of human AS were assessed. RESULTS: The expressions of MFG-E8 in murine and human AS cells were significantly higher than those in melanoma cells, macrophages and endothelial cells. Depletion of MFG-E8 in murine AS cells by siRNA significantly inhibited the formation of capillary-like structures and migration, but not proliferation. Administration of anti-MFG-E8 Ab significantly inhibited tumor growth and decreased the number of tumor-associated macrophages (TAMs) in AS tumors. Tumor size and the number of TAMs in human AS with high expression of MFG-E8 were significantly increased compared to those of AS with low expression of MFG-E8. Progression-free survival and overall survival time of the patients of AS with high expression of MFG-E8 were significantly shorter than those of AS with low expression of MFG-E8. CONCLUSIONS: AS-derived MFG-E8 might enhance tumor growth via angiogenesis and the induction of TAMs in autocrine/paracrine manner, and administration of anti-MFG-E8 Ab could be a therapeutic potential for AS.


Subject(s)
Antigens, Surface/metabolism , Hemangiosarcoma/pathology , Milk Proteins/metabolism , Neovascularization, Pathologic/pathology , Skin Neoplasms/pathology , Aged , Animals , Antibodies/administration & dosage , Antigens, Surface/genetics , Biopsy , Cell Line, Tumor/transplantation , Disease Models, Animal , Female , Gene Knockdown Techniques , Hemangiosarcoma/blood supply , Hemangiosarcoma/drug therapy , Human Umbilical Vein Endothelial Cells , Humans , Male , Mice , Milk Proteins/antagonists & inhibitors , Milk Proteins/genetics , Neovascularization, Pathologic/drug therapy , Pericytes , RAW 264.7 Cells , RNA, Small Interfering/metabolism , Skin/blood supply , Skin/pathology , Skin Neoplasms/blood supply , Skin Neoplasms/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...