Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Exp Dermatol ; 31(4): 582-585, 2022 04.
Article in English | MEDLINE | ID: mdl-35020224

ABSTRACT

The mechanism underlying giant congenital melanocytic nevus (GCMN) formation is not fully understood. According to recent research, NRAS gene mutation is the main driving factor in GCMN. Melanocytic precursor cells proliferate during the embryonic stage after acquiring NRAS mutations. However, why GCMN undergoes intense proliferation in the embryonic stage and then stops postnatally remains unknown. The current theory for this phenomenon is that the GCMN undergoes oncogene-induced senescence. However, there is not enough evidence to indicate that senescence induces growth arrest in GCMN. In this study, we hypothesized that the expression level of the NRAS gene changes dynamically during the development and differentiation of neural crest cells into melanocytes and that the NRAS expression level determines whether the cell proliferates or becomes quiescent.


Subject(s)
Nevus, Pigmented , Skin Neoplasms , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Humans , Melanocytes/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mutation , Nevus, Pigmented/congenital , Nevus, Pigmented/genetics , Skin Neoplasms/genetics
2.
J Reconstr Microsurg ; 34(1): 35-40, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28922668

ABSTRACT

BACKGROUND: Inadequate neovascularization is a major risk factor that can lead to subsequent necrosis of prefabricated flaps. Recent evidence indicates that transient receptor potential cation channel, subfamily V, member 4 (TRPV4) activates growth and remodeling of collateral arteries in ischemia tissues by responding to elevated fluid shear stress (FSS). Therefore, we evaluated whether TRPV4 could increase neovascularization in prefabricated flaps in a rat model. METHODS: Rat prefabricated skin flaps were created by ligating the right femoral vascular pedicle and implanting it underneath abdominal flaps. Thirty-six male Sprague-Dawley rats were randomly assigned to three groups with different solutions injected subcutaneously in the implantation site around the pedicle: injected with normal saline as the control group; injected with 4α-Phorbol 12,13-didecanoate (4αPDD), a specific TRPV4 activator, as the 4αPDD group; or injected with ruthenium red (RR), a TRPV-blocker, as the RR group. Neovascularization was evaluated by laser speckle contrast imaging (FLPI), histological staining, and enzyme-linked immunosorbent assay (ELISA) within two weeks. Afterwards, the abdominal island flaps were completely elevated and sutured back. The flap viability and survival area were examined on day 7. RESULTS: A larger area of flap survival, higher capillary densities, and higher von Willebrand factor (vWF) expression were observed in the 4αPDD group in comparison to those in the other two groups. The secretion of vascular endothelial growth factor (VEGF), but not basic fibroblast growth factor (bFGF), was significantly elevated in the 4αPDD group. CONCLUSION: Activation of TRPV4 using 4αPDD can significantly increase the survival of prefabricated flaps via neovascularization inducement, possibly through VEGF secretion enhancement. TRPV4 serves as a potential therapeutic neovascularization target in prefabricated flaps.


Subject(s)
Free Tissue Flaps/blood supply , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/physiology , Phorbol Esters/pharmacology , TRPV Cation Channels/metabolism , Animals , Disease Models, Animal , Femoral Artery , Graft Survival , Male , Rats , Rats, Sprague-Dawley , Plastic Surgery Procedures
3.
Ophthalmic Plast Reconstr Surg ; 33(5): 367-371, 2017.
Article in English | MEDLINE | ID: mdl-27755395

ABSTRACT

BACKGROUND: Conventional reconstructive methods fail to achieve satisfactory results in total eyelid defect cases. Vascularized composite tissue allotransplantation might provide both good appearance and function for these patients. The structure of the eyelid is exceptional because it simultaneously consists of skin, connective tissue, the striated muscle, fiber structure, aponeuroses, and mucosa. Thus, before clinical application of eyelid allotransplantation, more experiments are needed to clarify the impact of ischemia, immunal suppressive agents, and deinnervation effects on these sophisticated structures. We developed an heterotopic periorbital transplantation model in rats to facilitate further experiment in this field. METHODS: Twenty-five inbred male Lewis rats were used for anatomy study (n=10), and as donors or recipients of the operations (n=10). In the anatomy study, the vascular distribution and innervation to the periorbital unit was identified and recorded. Then, according to the anatomy study, 10 heterotopic transplantations and 2 transplantations with pedicle ligated were performed. The posterior facial vein and the external carotid artery are selected as the graft pedicle. All transplanted eyelids were assessed daily. Micro-CT scanning and hematoxylin and eosin staining of the grafts were performed 60 days after the operation. RESULTS: All recipients tolerated the operation well. All grafts without pedicles ligated survived and new hair growth was observed. All of the transplanted eyelids were pink and pliable during the entire observation period, and we did not observe any signs of arterial or venous occlusion. In the recipients with graft pedicle ligated, the grafts were necrosed and mummified within 4 to 5 days. MicroCT of the survived grafts showed good blood supply and histologic staining revealed normal histologic morphologies. CONCLUSIONS: Our study proved the anatomical feasibility of periorbital transplantation by establishing a heterotopic transplantation model, which might facilitate future eyelid allotransplantation-related experiments.


Subject(s)
Blepharoplasty/methods , Eyelids/surgery , Models, Anatomic , Vascularized Composite Allotransplantation/methods , Animals , Feasibility Studies , Male , Models, Animal , Rats , Rats, Inbred Lew
4.
Cell Physiol Biochem ; 40(1-2): 207-218, 2016.
Article in English | MEDLINE | ID: mdl-27855376

ABSTRACT

BACKGROUND: Keloids are fibroproliferative scars that develop as a result of a dysregulated wound healing process; however, the molecular mechanisms of keloid pathogenesis remain unclear. Keloids are characterized by the ability to spread beyond the original boundary of the wound, and they represent a significant clinical challenge. Previous work from our group suggested that growth differentiation factor (GDF)-9 plays a role in the invasive behavior of keloids. Here, we examined the involvement of GDF-9 in keloid formation and spread and elucidated a potential underlying mechanism. METHODS: The expression of GDF-9, cyclooxygenase (COX)-2, vascular epidermal growth factor (VEGF)-C, matrix metalloprotease (MMP)-2, MMP-9, transforming growth factor (TGF)-ß1, and the related signaling pathway components in human keloid tissues or keloid fibroblasts (kFBs) was monitored by qRT-PCR and western blot. A series of overexpression and silencing experiments in normal and keloid fibroblasts were used to modify the expression of GDF-9. The effects of GDF-9 on kFB proliferation and migration were assessed using the CCK-8, cell cycle and scratch wound healing assays. RESULTS: GDF-9 promotes fibroblast proliferation and migration. GDF-9 silencing in kFBs decreased cell proliferation, blocked cell cycle progression, downregulated the angiogenic markers COX-2 and VEGF-C, and downregulated MMP-2 and MMP-9 expression, whereas it had no effect on the levels of TGF-ß1. GDF-9 silencing significantly inhibited Smad2 and Smad3 phosphorylation in kFBs. CONCLUSIONS: GDF-9 promotes the proliferation and migration of kFBs via a mechanism involving the Smad2/3 pathway.


Subject(s)
Cell Movement , Fibroblasts/pathology , Growth Differentiation Factor 9/metabolism , Keloid/pathology , Signal Transduction , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Adult , Cell Proliferation , Cyclooxygenase 2/metabolism , Female , Fibroblasts/metabolism , Gene Silencing , Growth Differentiation Factor 9/genetics , Humans , Keloid/enzymology , Keloid/genetics , Male , Middle Aged , Up-Regulation/genetics , Vascular Endothelial Growth Factor C/genetics , Vascular Endothelial Growth Factor C/metabolism , Young Adult
5.
Virol J ; 12: 10, 2015 Feb 03.
Article in English | MEDLINE | ID: mdl-25645259

ABSTRACT

BACKGROUND: After the 1968 H3N2 pandemic emerged in humans, H3N2 influenza viruses continuously circulated and evolved in nature. An H3N2 variant was circulating in humans in the 1990s and subsequently introduced into the pig population in the 2000s. This virus gradually became the main subtype of swine influenza virus worldwide. However, there were no reports of infections in dogs with this virus. FINDINGS: In 2013, 35 nasal swabs from pet dogs were positive for Influenza A virus by RT-PCR. Two viruses were isolated and genetically characterized. In the phylogenetic trees of all gene segments, two H3N2 canine isolates clustered with Moscow/10/99 and most H3N2 swine influenza viruses. These results indicated that two H3N2 CIVs possessed high homology with human/swine influenza viruses, which at the same time exhibited some amino acid substitutions in NA, polymerase basic protein 1 (PB1), and nucleoprotein (NP), which probably were related to the interspecies transmission. CONCLUSIONS: These two viruses share the highest homology with swine H3N2, Moscow/99-like viruses, which indicated that these viruses might originate from swine viruses.


Subject(s)
Dog Diseases/virology , Influenza A Virus, H3N2 Subtype/classification , Influenza A Virus, H3N2 Subtype/isolation & purification , Orthomyxoviridae Infections/veterinary , Animals , China , Cluster Analysis , Dogs , Influenza A Virus, H3N2 Subtype/genetics , Molecular Sequence Data , Mutation, Missense , Nasal Mucosa/virology , Orthomyxoviridae Infections/virology , Pets , Phylogeny , RNA, Viral/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , Sequence Homology , Viral Proteins/genetics
6.
J Surg Res ; 199(2): 707-17, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26081004

ABSTRACT

BACKGROUND: Finger allotransplantation is a promising treatment for severe finger destruction. However, more research is required to decrease the risks of this procedure to a level at which the clinical use of this non-life-saving procedure is justified. A proper animal model is essential for the required experiments. METHODS: In this article, we established a toe transplantation model based on anatomic studies. A tapered dose of Cyclosporine A (CsA) was used as an immunosuppressive therapy in the Brown Norway-to-Lewis allotransplantation experimental group, whereas isotransplantation or allotransplantation without treatment or with ligated pedicles was performed on the control groups. Recipients were assessed daily after operation for any signs of rejection and complications. On postoperative day 90, skin graft test was used to test the level of donor-specific tolerance in the recipients. On postoperative day 120, x-rays and micro-computed tomographies were performed for bone morphology evaluation. The chimerism in the recipient peripheral blood, lymph node, spleen, and thymus was tested by flow cytometry and immunohistochemical staining. And histologic study of the toe grafts and skin grafts were carried out. RESULTS: The blood supply of the toe graft was confirmed, and accordingly, transplantations were performed. The isografts survived indefinitely. The allografts with ligated pedicles experienced necrosis within 5 d. The allografts without treatment exhibited necrosis within 14 d. Forty percent, 20%, and 40% of the allografts associated with the CsA treatment experienced severe rejection, mild rejection, and nonrejection, according to gross graft appearance, respectively. Skin grafting tests showed three different types of results. X-rays and micro-computed tomographies reveal nearly normal bone morphologies in the bone structures of all surviving animals with grafts. Low levels of donor-specific chimerism were detected in the peripheral blood samples. Spleen, lymph node, and thymus chimerism were also confirmed in the long-term surviving animals with allografts. Histologic evaluation of the long-term survivals revealed similar graft morphologies in the isografts and the nonrejected allografts, inflammatory cell infiltration in the mildly rejected allografts, and degraded cutaneous appendages in the severely rejected allografts. CONCLUSIONS: We established a toe allotransplantation model. Moreover, the low rate of chimerism did not introduce specific tolerance, which might explain the high rejection rate. This model might facilitate future research in finger allotransplantation.


Subject(s)
Models, Animal , Toes/transplantation , Animals , Chimerism , Male , Radiography , Rats, Inbred BN , Skin Transplantation , Transplantation, Homologous , Transplants/diagnostic imaging , Transplants/pathology
7.
Cancer Gene Ther ; 30(10): 1390-1402, 2023 10.
Article in English | MEDLINE | ID: mdl-37488294

ABSTRACT

Malignant peripheral nerve sheath tumors (MPNSTs) are aggressive soft-tissue sarcomas characterized by poor prognosis and low drug response rates. Traditional chemo/radiotherapies show only mild benefits for patients with MPNSTs, and no targeted therapy is available in the clinic. A better understanding of the molecular background of MPNSTs is critical for the development of effective targeted therapies. Forkhead box M1 (FOXM1) has been implicated in the progression of many human malignancies, though its role in MPNSTs is unclear. In this study, using four Gene Expression Omnibus (GEO) datasets and a tissue microarray, we demonstrated that FOXM1 upregulation was associated with poor prognosis in patients with MPNSTs. FOXM1 overexpression and knockdown regulated the proliferation and colony formation of MPNST cells. Using bioinformatics analysis and luciferase reporter assays, we identified NUF2 as a direct downstream target of FOXM1. Both in vitro and in vivo experiments demonstrated that the induction of MPNST cell proliferation by FOXM1 was dependent on elevated NUF2 expression, as NUF2 knockdown abolished the FOXM1-induced proliferation of MPNST cells. Our study showed that the FOXM1-NUF2 axis mediates human MPNST progression and could be a potential therapeutic target.


Subject(s)
Forkhead Box Protein M1 , Nerve Sheath Neoplasms , Neurofibromatosis 1 , Humans , Cell Cycle Proteins , Cell Proliferation , Forkhead Box Protein M1/genetics , Nerve Sheath Neoplasms/genetics , Nerve Sheath Neoplasms/metabolism , Nerve Sheath Neoplasms/pathology , Neurofibromatosis 1/complications , Neurofibromatosis 1/genetics , Neurofibromatosis 1/pathology , Neurofibrosarcoma/complications
8.
Cell Oncol (Dordr) ; 46(5): 1399-1413, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37086345

ABSTRACT

BACKGROUND: Malignant peripheral nerve sheath tumors (MPNSTs) are aggressive sarcomas that typically develop in the setting of neurofibromatosis type 1 (NF1) and cause significant morbidity. Conventional therapies are often ineffective for MPNSTs. Ribonucleotide reductase subunit M2 (RRM2) is involved in DNA synthesis and repair, and is overexpressed in multiple cancers. However, its role in NF1-associated MPNSTs remains unknown. Our objective was to determine the therapeutic and prognostic potential of RRM2 in NF1-associated MPNSTs. METHODS: Identification of hub genes was performed by using NF1-associated MPNST microarray datasets. We detected RRM2 expression by immunochemical staining in an MPNST tissue microarray, and assessed the clinical and prognostic significance of RRM2 in an MPNST cohort. RRM2 knockdown and the RRM2 inhibitor Triapine were used to assess cell proliferation and apoptosis in NF1-associated MPNST cells in vitro and in vivo. The underlying mechanism of RRM2 in NF1-associated MPNST was revealed by transcriptome analysis. RESULTS: RRM2 is a key hub gene and its expression is significantly elevated in NF1-associated MPNST. We revealed that high RRM2 expression accounted for a larger proportion of NF1-associated MPNSTs and confirmed the correlation of high RRM2 expression with poor overall survival. Knockdown of RRM2 inhibited NF1-associated MPNST cell proliferation and promoted apoptosis and S-phase arrest. The RRM2 inhibitor Triapine displayed dose-dependent inhibitory effects in vitro and induced significant tumor growth reduction in vivo in NF1-associated MPNST. Analysis of transcriptomic changes induced by RRM2 knockdown revealed suppression of the AKT-mTOR signaling pathway. Overexpression of RRM2 activates the AKT pathway to promote NF1-associated MPNST cell proliferation. CONCLUSIONS: RRM2 expression is significantly elevated in NF1-associated MPNST and that high RRM2 expression correlates with poorer outcomes. RRM2 acts as an integral part in the promotion of NF1-associated MPNST cell proliferation via the AKT-mTOR signaling pathway. Inhibition of RRM2 may be a promising therapeutic strategy for NF1-associated MPNST.


Subject(s)
Neurofibromatosis 1 , Neurofibrosarcoma , Humans , Neurofibromatosis 1/complications , Neurofibromatosis 1/genetics , Neurofibromatosis 1/pathology , Neurofibrosarcoma/complications , Neurofibrosarcoma/pathology , Neurofibrosarcoma/therapy , Proto-Oncogene Proteins c-akt/metabolism , Prognosis , TOR Serine-Threonine Kinases/metabolism
9.
Front Pediatr ; 10: 784660, 2022.
Article in English | MEDLINE | ID: mdl-35281247

ABSTRACT

Background: Large congenital melanocytic nevus (LCMN) is a rare skin disease that deeply affects an individual's appearance, may influence patients' self-evaluation and social relationships, and further affects their quality of life (QoL). The Skindex-29 and 36-Item Short Form Survey (SF-36) are valid instruments used to evaluate QoL specifically. It is necessary to assess the QoL of patients with LCMN and summarize potentially impactful factors to help people understand LCMN patients and assist doctors in offering professional advice. Methods: Twenty-five patients were recruited from Shanghai Ninth People's Hospital from July 1st, 2019, to March 31st, 2021. Both males and females were included, and the age groups were divided into infants (0-6 y), children (7-12 y), teenagers (13-17 y) and youths (18-45 y). The Skindex-29 and SF-36 were applied as questionnaires for the assessment of QoL. Clinical information was acquired by physical examination. Results: QoL in patients with LCMN was diminished, especially in the emotional aspect. However, different genders, ages and distribution patterns of LCMN did not significantly influence QoL, but the patterns of "Bonce" and "Body" affected QoL the most and the severest. The results of Skindex-29 and SF-36 were consistent in that LCMN mainly reduced QoL from an emotional perspective. Conclusions: This research shows that LCMN has the strongest impact on patients' emotional wellbeing but weakly influences the whole fettle of QoL. The gender, age and distribution patterns of lesions all have no direct effect on QoL, although a larger proportion of LCMNs probably insinuates worse QoL. Even though patients with LCMN show better QoL than those with other visible skin conditions, their general mental health still requires ample attention from surroundings and professional doctors.

10.
Dis Markers ; 2022: 8114731, 2022.
Article in English | MEDLINE | ID: mdl-35722625

ABSTRACT

Cutaneous malignant melanoma (CMM) is the most deadly skin cancer worldwide. Despite advances in the treatments of CMM, its incidence and mortality rates are still increasing. N6-methyladenosine (m6A) is the most common form of RNA modification and has attracted increasing interest in cancer initiation and progression. However, the role of m6A regulators in CMM and their correlation with prognosis remain elusive. Here, we demonstrated that by applying consensus clustering, all CMM patient cases can be divided into two clusters based on overall expression levels of 25 m6A genes. We systematically analyzed the prognostic value of the 25 m6A RNA methylation regulators in CMM and found that ELAVL1, ABCF1, and IGF2BP1 yield the highest scores for predicting the prognosis of CMM. Accordingly, we derived a risk signature consisting of three selected m6A genes as an independent prognostic marker for CMM and validated our findings with data derived from a different CMM cohort. Next, we determined that CNVs in m6A genes had a significant negative impact on patient survival. The mRNA expression levels of m6A genes were correlated with CNV mutation. Moreover, in the selected three risk signature m6A regulators, GSEA analysis showed that they were closely correlated with inflammation and immune pathways. TME analysis proved that m6A gene expressions were negatively correlated with immune cell infiltration. In conclusion, m6A regulators are vital participants in CMM pathology; and ELAVL1, ABCF1, and IGF2BP1 mRNA levels are valuable factors for prognosis prediction and treatment strategy development.


Subject(s)
Melanoma , Skin Neoplasms , ATP-Binding Cassette Transporters , Computational Biology , Humans , Melanoma/genetics , Melanoma/pathology , Prognosis , RNA, Messenger/metabolism , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Melanoma, Cutaneous Malignant
11.
Pigment Cell Melanoma Res ; 35(3): 356-368, 2022 05.
Article in English | MEDLINE | ID: mdl-35218152

ABSTRACT

Treatments for giant congenital melanocytic nevi (GCMN) are extremely limited. Thus, there is an urgent need for development of relevant targeted therapies. However, current lack of preclinical cell models restricts progress in GCMN research. In this study, we aimed to establish and characterize an immortalized GCMN cell line. GCMN cells were successfully immortalized by means of lentivirus-mediated simian virus 40 large T transfection. The immortalized GNC cell line (ImGNC) showed lower proliferation rate and higher melanin content than primary melanocytes. Expression levels of the differentiation gene MITF and stemness genes TWIST1, SNAI1, and FOXD3 were elevated in ImGNCs; however, the established ImGNC cell line was immortalized but not transformed. Sanger sequencing detected the heterozygous NRASQ61K mutation in ImGNCs, but not the BRAFV600E mutation. Despite carrying the NRASQ61K allele, ImGNCs demonstrated suppressed MAPK activation and elevated PI3K/Akt activation, as compared with primary melanocytes. Drug sensitivity analysis showed that ImGNCs are more sensitive to PI3K/Akt and Bcl-2 inhibitors than to MEK or ERK inhibitors. Unlike the proliferation-inhibiting effect of PI3K/Akt inhibitors, the Bcl-2 inhibitor navitoclax promptly promoted apoptosis in ImGNCs. Considering the low proliferation characteristics of GCMN in vivo, Bcl-2 may be a potential therapeutic target that warrants further research.


Subject(s)
Antineoplastic Agents , Nevus, Pigmented , Cell Line , Humans , Nevus, Pigmented/congenital , Nevus, Pigmented/genetics , Phosphatidylinositol 3-Kinases , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins c-bcl-2 , Skin Neoplasms
13.
Burns ; 46(2): 377-385, 2020 03.
Article in English | MEDLINE | ID: mdl-31852618

ABSTRACT

Skin grafts are widely used in plastic and reconstructive surgery. Increasing the early vascularization of skin grafts is a key factor in improving skin grafting. In this study, we use platelet-rich plasma gel as an adipose-derived stem cell scaffold to assist the growth of rat skin grafts. ADSCs were successfully isolated and seeded into the PRP gel. Using a Lewis rat model, we found the PRP gel + ADSCs significantly improved the properties of the transplanted skin grafts, increased the skin thickness and improved the collagen arrangement. PRP gel + ADSCs promoted skin neovascularization by elevating the expression of the vascularization factors VEGF, BFGF and PDGFB. Taken together, our study indicated that ADSCs combined with PRP have a potentiation effect on improving skin grafts by promoting angiogenesis, providing an innovative approach and a theoretical basis for its clinical application.


Subject(s)
Gels , Mesenchymal Stem Cell Transplantation/methods , Neovascularization, Physiologic/genetics , Platelet-Rich Plasma , Skin Transplantation/methods , Skin/pathology , Tissue Scaffolds , Animals , Elastic Modulus , Fibroblast Growth Factor 2/genetics , Luminescent Measurements , Mesenchymal Stem Cells , Microscopy, Electron, Scanning , Proto-Oncogene Proteins c-sis/genetics , RNA, Messenger/metabolism , Rats , Rats, Inbred Lew , Skin/metabolism , Skin Physiological Phenomena , Vascular Endothelial Growth Factor A/genetics
14.
Exp Ther Med ; 19(1): 313-318, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31853305

ABSTRACT

Giant congenital melanocytic nevi (GCMN) place a heavy psychological burden on patients due to their poor cosmetic appearance. The histopathological characteristics of GCMN have remained largely elusive. The present study investigated the histopathological characteristics of GCMN in association with their clinical appearance. A total of 98 patients diagnosed with GCMN were included in the present study and their clinical features were collected from their records. Lesion specimens were obtained and stained for histopathological analysis. Regarding the microscopic appearance of GCMN, nevi cells in the whole dermis exhibited different patterns than those in healthy tissues. Most GCMN cases featured a sub-epidermal non-involvement zone, which implies an early occurrence in embryo development. Darker nevi exhibited a higher density of infiltrated nevi cells and more pigment deposition; this appears to induce a poor skin texture. Chemical peeling and laser therapy only partly removes pigment particles and nevi cells in the upper portion of the dermis. The clinical features of GCMN are associated with the histopathological characteristics, and non-surgical therapy cannot remove the nevus cells in the deep dermis.

15.
Am J Transl Res ; 10(4): 1184-1194, 2018.
Article in English | MEDLINE | ID: mdl-29736211

ABSTRACT

Most giant congenital melanocytic nevi (GCMN) exhibit an activating mutation in NRAS. Constitutive activation of the RAS-ERK signaling pathway induces proliferation in nevus cells and plays a pivotal role in melanoma development. In this study, we studied the efficacy of RAS-ERK pathway targeted therapy in GCMN. We isolated nevus cells from GCMN (GNCs) and compared the morphology of GNCs with normal melanocytes and the A375 melanoma cell line. Proliferation curves of GNCs and A375 cells were determined using Cell Counting Kit-8 assays. Cell cycle distribution was measured using flow cytometry. The RAS-ERK pathway inhibitors Vemurafenib and Trametinib, which are used in melanoma therapy, were applied. After inhibitor treatment, GNCs were analyzed for apoptosis and the protein expression of ERK, p-ERK, P38, p-P38 and P53. We found that compared with A375 cells, the cultured GNCs exhibited a higher G1 phase population and a lower proliferation rate. Both Vemurafenib and Trametinib treatment induced GNCs apoptosis in a dose-dependent manner, with Vemurafenib having a stronger effect. With inhibitor treatment, ERK activation was greatly suppressed, while the expression of p-P38 exhibited no obvious change. Vemurafenib treatment also increased the level of P53 protein in GNCs. These findings suggested that Vemurafenib treatment may be a potential therapeutic strategy for treatment of GCMN via targeting of the RAS-ERK pathway.

16.
Emerg Microbes Infect ; 7(1): 85, 2018 May 16.
Article in English | MEDLINE | ID: mdl-29765037

ABSTRACT

Considered a "mixing vessel" for influenza viruses, pigs can give rise to new influenza virus reassortants that can threaten humans. During our surveillance of pigs in Guangxi, China from 2013 to 2015, we isolated 11 H1N1 and three H3N2 influenza A viruses of swine origin (IAVs-S). Out of the 14, we detected ten novel triple-reassortant viruses, which contained surface genes (hemagglutinin and neuraminidase) from Eurasian avian-like (EA) H1N1 or seasonal human-like H3N2, matrix (M) genes from H1N1/2009 pandemic or EA H1N1, nonstructural (NS) genes from classical swine, and the remaining genes from H1N1/2009 pandemic. Mouse studies indicate that these IAVs-S replicate efficiently without prior adaptation, with some isolates demonstrating lethality. Notably, the reassortant EA H1N1 viruses with EA-like M gene have been reported in human infections. Further investigations will help to assess the potential risk of these novel triple-reassortant viruses to humans.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/genetics , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H3N2 Subtype/genetics , Neuraminidase/genetics , Reassortant Viruses/genetics , Reassortant Viruses/isolation & purification , Animals , Cell Line , China , Disease Outbreaks/veterinary , Dogs , Female , Influenza A Virus, H1N1 Subtype/isolation & purification , Influenza A Virus, H3N2 Subtype/isolation & purification , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred BALB C , Phylogeny , Swine , Swine Diseases/virology
17.
Exp Ther Med ; 13(1): 55-62, 2017 Jan.
Article in English | MEDLINE | ID: mdl-28123468

ABSTRACT

Bone marrow-derived mesenchymal stem cell (BM-MSC) transplantation has been demonstrated to be an effective way of augmenting angiogenesis of ischemic tissue. The low oxygen conditions in ischemic tissue directly affect the biological behavior of engrafted cells. However, to date, the mechanism through which hypoxia regulates self-renewal, differentiation and paracrine function of BM-MSCs remains unclear. Clarification of this mechanism would be beneficial to the use of stem cell-based therapy. The PI3K/AKT pathway has been extensively investigated for its role in cell proliferation, cell transformation, paracrine function and angiogenesis. The present study aimed to analyze the role of PI3K/AKT pathway in hypoxia-induced proliferation of BM-MSCs and their differentiation into endothelial cells in vitro by the application of LY294002, a PI3K/AKT pathway inhibitor, with cells cultured in normoxia serving as a control. The results showed that rat BM-MSCs at passage 3 and 4 displayed only few phenotypical differences in the expression of surface antigens as detected by flow cytometry. When compared with the cells treated in normoxia, the proliferation of BM-MSCs in hypoxia was promoted, a greater number of cells expressed CD31 and a higher expression of vascular endothelial growth factor was observed after culture in hypoxic conditions. However, by inhibiting with LY294002, these changes induced by hypoxia were partly inhibited. In conclusion, the present study showed that the PI3K/AKT pathway served an important role in hypoxia-enhanced in vitro proliferation of BM-MSCs and their differentiation into endothelial cells and paracrine vascular endothelial growth factor.

18.
Sci Rep ; 7(1): 1607, 2017 05 09.
Article in English | MEDLINE | ID: mdl-28487509

ABSTRACT

Temporary ectopic implantation has been performed in clinical practice to salvage devascularized amputated tissues for delayed replantation purpose. In this study, we established a series of segmental forelimb ectopic implantation models in rats, including forelimb, forearm, forepaw, digit, and double forelimbs, to mimic the clinical context. Time of amputated limbs harvesting in donors and ectopic implantation process in recipients were recorded. Survival time and mortalities of recipients were also recorded. Sixty days after ectopic implantation, a full-field laser perfusion imager (FLPI) was used to detect the blood flow of amputated limbs and micro-CT imaging was used to examine bone morphological changes. Histological sections of amputated limbs were stained with hematoxylin and eosin to evaluate pathological changes. Implanted amputated limbs in all models achieved long term survival and there were no obvious morphological and histological changes were found according to results of micro-CT and histology study. Thus, a series of rat segmental forelimb temporary ectopic implantation models have been well established. To our knowledge, this is the first rodent animal model related to forelimb temporary ectopic implantation. These models might facilitate further research related to salvage, reconstruction and better aesthetic and functional outcome of upper extremity/digit in temporary ectopic implantation scenario.


Subject(s)
Forelimb/physiology , Prosthesis Implantation , Amputation, Surgical , Animals , Biopsy , Forelimb/blood supply , Forelimb/pathology , Male , Operative Time , Rats, Inbred Lew , Time Factors
19.
Plast Reconstr Surg ; 139(2): 376-390, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28121871

ABSTRACT

BACKGROUND: Keloid scarring impairs patients' quality of life, and although many therapeutic strategies have been developed, most remain unsatisfactory because of limited understanding of the mechanisms underlying keloid development. METHODS: A microarray gene expression data set from keloid tissue was acquired from the Gene Expression Omnibus. Differentially expressed genes in fibroblasts and keratinocytes underwent functional annotation and pathway analysis. Weighted gene coexpression network analysis was applied to identify the gene targets of keloid scars within differentially expressed genes. Modules and hub genes for keloids were identified. Enrichment analysis was undertaken to verify the modules' and hub genes' relationship with keloids. RESULTS: Enrichment analysis and pathway analysis showed gene ontology terms and pathways related to keloids. Each cell type generated three modules in weighted gene coexpression network analysis, with one module most related to keloids. Enrichment analysis showed that the modules concerned are enriched with terms related to keloids. Three hub genes were selected for fibroblasts and keratinocytes, and their relationship to keloids was verified. Immunohistochemical staining verified expression change of some hub genes. CONCLUSIONS: This is the first study to describe the gene networks underlying keloids. Modules and hub genes generated in the present study are highly related to keloids and may identify novel therapeutic targets for treatment of keloids. CLINICAL QUESTION/LEVEL OF EVIDENCE: Therapeutic, V.


Subject(s)
Gene Expression , Gene Regulatory Networks , Keloid/genetics , Humans , Microarray Analysis
20.
Oncotarget ; 7(50): 82074-82084, 2016 Dec 13.
Article in English | MEDLINE | ID: mdl-27738338

ABSTRACT

Alopecia is a dermatological condition with limited therapeutic options. Only two drugs, finasteride and minoxidil, are approved by FDA for alopecia treatment. However, little is known about the differences in adverse effects between these two drugs. We examined the clinical reports submitted to the FDA Adverse Event Reporting System (FAERS) from 2004 to 2014. For both female and males, finasteride was found to be more associated with reproductive toxicity as compared to minoxidil. Among male alopecia cases, finasteride was significantly more concurrent with several forms of sexual dysfunction. Among female alopecia cases, finasteride was significantly more concurrent with harm to fetus and disorder of uterus. In addition, drug-gene network analysis indicated that finasteride could profoundly disturb pathways related to sex hormone signaling and oocyte maturation. These findings could provide clues for subsequent toxicological research. Taken together, this analysis suggested that finasteride could be more liable to various reproductive adverse effects. Some of these adverse effects have yet to be warned in FDA-approved drug label. This information can help improve the treatment regimen of alopecia and post-marketing regulation of drug products.


Subject(s)
5-alpha Reductase Inhibitors/adverse effects , Alopecia/drug therapy , Drug-Related Side Effects and Adverse Reactions/etiology , Finasteride/adverse effects , Minoxidil/adverse effects , Reproduction/drug effects , Adverse Drug Reaction Reporting Systems , Alopecia/diagnosis , Alopecia/physiopathology , Drug-Related Side Effects and Adverse Reactions/metabolism , Drug-Related Side Effects and Adverse Reactions/pathology , Drug-Related Side Effects and Adverse Reactions/physiopathology , Female , Fetus/drug effects , Fetus/metabolism , Fetus/pathology , Gene Expression Regulation/drug effects , Gene Regulatory Networks/drug effects , Hair/drug effects , Hair/growth & development , Humans , Male , Risk Assessment , Risk Factors , Sex Factors , United States , United States Food and Drug Administration , Uterus/drug effects , Uterus/metabolism , Uterus/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL