1
|
Ren H, Zhao Q, Wang N, Yuan X, Song R, Wen Q, Zhao Y. Melanoma-inhibiting activity promotes the migration and odontoblastic differentiation of stem cells of apical papilla. Arch Oral Biol 2025; 169:106109. [PMID: 39427554 DOI: 10.1016/j.archoralbio.2024.106109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 10/11/2024] [Accepted: 10/13/2024] [Indexed: 10/22/2024]
Abstract
OBJECTIVE Melanoma Inhibitory Activity (MIA) has been predominantly studied in the context of melanoma and cartilage development. However, its role in dental pulp development and stem cell behavior remains largely unexplored. This study investigates the expression pattern of MIA in dental pulp tissues and its potential role in the proliferation, migration, and odontoblastic differentiation of stem cells from the apical papilla (SCAPs). DESIGN MIA expression in human pulp tissue was demonstrated by immunohistochemistry. SCAPs were cultured in normal and mineralization induction media, with MIA levels monitored via RT-qPCR and Western blot. Cell proliferation was evaluated using the CCK8 assay, while transwell and cell scratch assays were conducted to examine cell migration. The effect of MIA on odontoblastic differentiation was examined by qRT-PCR, Alkaline phosphatase activity assay, and Western blot. siRNA was used to knock down MIA to investigate its effect. A mouse subcutaneous implantation model was used to assess whether MIA promotes odontoblastic differentiation in vivo. RESULTS MIA expression was observed in the papilla and odontoblasts layer of the developing pulp. In vitro, MIA expression increased during SCAPs differentiation and was found to significantly enhance migration, and odontoblastic differentiation but not proliferation. Gene knockdown experiments confirmed MIA's pivotal role in promoting SCAPs migration and differentiation. In vivo, MIA facilitated the formation of dentin-like structures and enhanced pulp-dentin complex regeneration. CONCLUSION MIA plays a crucial role in SCAPs' migration and differentiation, suggesting its potential application in pulp-dentin regeneration therapies. Further studies are required to fully elucidate the underlying mechanisms.
Collapse
Affiliation(s)
- Huihui Ren
- Department of Pediatric Dentistry, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, No.22, Zhongguancun South Avenue, Haidian District, Beijing 100081, PR China
| | - Qingxuan Zhao
- Department of Pediatric Dentistry, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, No.22, Zhongguancun South Avenue, Haidian District, Beijing 100081, PR China
| | - Nan Wang
- Department of Stomatology, Peking University Third Hospital, Beijing 100191, PR China
| | - Xiaojing Yuan
- Department of Pediatric Dentistry, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, No.22, Zhongguancun South Avenue, Haidian District, Beijing 100081, PR China
| | - Rui Song
- Department of Pediatric Dentistry, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, No.22, Zhongguancun South Avenue, Haidian District, Beijing 100081, PR China
| | - Quan Wen
- First Clinical Division, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, 37A Xishiku Street, Xicheng District, Beijing 100034, PR China.
| | - Yuming Zhao
- Department of Pediatric Dentistry, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, No.22, Zhongguancun South Avenue, Haidian District, Beijing 100081, PR China.
| |
Collapse
|
2
|
Akkour K, Alanazi IO, Alfadda AA, Masood A, Alhalal H, Joy SS, Bassi A, Alshehri E, Alwehaibi MA, Arafah M, Benabdelkamel H. Plasma-based proteomic profiling identifies the distinct regulation of proteins in hyperplasia and endometrial cancer. BMC Cancer 2024; 24:752. [PMID: 38902713 PMCID: PMC11191338 DOI: 10.1186/s12885-024-12522-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 06/14/2024] [Indexed: 06/22/2024] Open
Abstract
BACKGROUND Among gynaecological malignancies, endometrial cancer (EC) is the most prevalent type of uterine cancer affecting women. This study explored the proteomic profiles of plasma samples obtained from EC patients, those with hyperplasia (Hy), and a control group (CO). A combination of techniques, such as 2D-DIGE, mass spectrometry, and bioinformatics, including pathway analysis, was used to identify proteins with modified expression levels, biomarkers and their associated metabolic pathways in these groups. METHODS Thirty-four patients, categorized into three groups-10 with EC, 12 with Hy, and 12 CO-between the ages of 46 and 75 years old were included in the study. Untargeted proteomic analysis was carried out using two-dimensional difference in gel electrophoresis (2D-DIGE) coupled with matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF-MS). RESULTS In all three groups, 114 proteins that were significantly (p ≤ 0.05 and fold change ≥ 1.5) altered were successfully identified using peptide mass fingerprints (PMFs). Compared with those in the control group (CO), the EC samples had 85 differentially expressed proteins (39 upregulated and 46 downregulated), and in the Hy group, 81 proteins were dysregulated (40 upregulated and 41 downregulated) compared to those in the CO group, while 33 proteins exhibited differential regulation (12 upregulated and 21 downregulated) in the EC plasma samples compared to those in the Hy group. Vitamin D binding protein and complement C3 distinguished Hy and EC from CO with the greatest changes in expression. Among the differentially expressed proteins identified, enzymes with catalytic activity represented the largest group (42.9%). In terms of biological processes, most of the proteins were involved in cellular processes (28.8%), followed by metabolic processes (16.7%). STRING analysis for protein interactions revealed that the significantly differentially abundant proteins in the three groups are involved in three main biological processes: signalling of complement and coagulation cascades, regulation of insulin-like growth factor (IGF) transport and uptake by insulin-like growth factor binding proteins (IGFBPs), and plasma lipoprotein assembly, remodelling, and clearance. CONCLUSION The identified plasma protein markers have the potential to serve as biomarkers for differentiating between EC and Hy, as well as for early diagnosis and monitoring of cancer progression.
Collapse
Affiliation(s)
- Khalid Akkour
- Obstetrics and Gynecology Department, College of Medicine, King Saud University, Riyadh, 11461, Saudi Arabia
| | - Ibrahim O Alanazi
- Healthy Aging Research Institute, King Abdulaziz City for Science and Technology (KACST), Health Sector, Riyadh, 11442, Saudi Arabia
| | - Assim A Alfadda
- Proteomics Resource Unit, Obesity Research Center, College of Medicine, King Saud University, Riyadh, 11461, Saudi Arabia
- Department of Medicine, College of Medicine, King Saud University, Riyadh, 11461, Saudi Arabia
| | - Afshan Masood
- Proteomics Resource Unit, Obesity Research Center, College of Medicine, King Saud University, Riyadh, 11461, Saudi Arabia
| | - Hani Alhalal
- Obstetrics and Gynecology Department, College of Medicine, King Saud University, Riyadh, 11461, Saudi Arabia
| | - Salini Scaria Joy
- Proteomics Resource Unit, Obesity Research Center, College of Medicine, King Saud University, Riyadh, 11461, Saudi Arabia
- Strategic Center for Diabetes Research, College of Medicine, King Saud University, Riyadh, 11461, Saudi Arabia
| | - Ali Bassi
- Obstetrics and Gynecology Department, College of Medicine, King Saud University, Riyadh, 11461, Saudi Arabia
| | - Eman Alshehri
- Obstetrics and Gynecology Department, College of Medicine, King Saud University, Riyadh, 11461, Saudi Arabia
| | - Moudi A Alwehaibi
- Proteomics Resource Unit, Obesity Research Center, College of Medicine, King Saud University, Riyadh, 11461, Saudi Arabia
| | - Maria Arafah
- Department of Pathology, College of Medicine, King Saud University, King Saud University Medical City, Riyadh, 11461, Saudi Arabia
| | - Hicham Benabdelkamel
- Proteomics Resource Unit, Obesity Research Center, College of Medicine, King Saud University, Riyadh, 11461, Saudi Arabia.
| |
Collapse
|
3
|
Cao J, Ma X, Zhang G, Hong S, Ma R, Wang Y, Yan X, Ma M. Prognostic analyses of genes associated with anoikis in breast cancer. PeerJ 2023; 11:e15475. [PMID: 37842046 PMCID: PMC10576492 DOI: 10.7717/peerj.15475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 05/08/2023] [Indexed: 10/17/2023] Open
Abstract
Breast cancer (BRCA) is the most diagnosed cancer worldwide and is responsible for the highest cancer-associated mortality among women. It is evident that anoikis resistance contributes to tumour cell metastasis, and this is the primary cause of treatment failure for BRCA. However, anoikis-related gene (ARG) expression profiles and their prognostic value in BRCA remain unclear. In this study, a prognostic model of ARGs based on The Cancer Genome Atlas (TCGA) database was established using a least absolute shrinkage and selection operator analysis to evaluate the prognostic value of ARGs in BRCA. The risk factor graph demonstrated that the low-risk group had longer survival than the high-risk group, implying that the prognostic model had a good performance. We identified 11 ARGs that exhibited differential expression between the two risk groups in TCGA and Gene Expression Omnibus databases. Through Gene Ontology and Kyoto Encyclopaedia of Genes and Genomes enrichment analyses, we revealed that the screened ARGs were associated with tumour progression and metastasis. In addition, a protein-protein interaction network showed potential interactions among these ARGs. Furthermore, gene set enrichment analysis suggested that the Notch and Wnt signalling pathways were overexpressed in the high-risk group, and gene set variation analysis revealed that 38 hallmark genes differed between the two groups. Moreover, Kaplan-Meier survival curves and receiver operating characteristic curves were used to identify five ARGs (CD24, KRT15, MIA, NDRG1, TP63), and quantitative polymerase chain reaction was employed to assess the differential expression of these ARGs. Univariate and multivariate Cox regression analyses were then performed for the key ARGs, with the best prediction of 3 year survival. In conclusion, ARGs might play a crucial role in tumour progression and serve as indicators of prognosis in BRCA.
Collapse
Affiliation(s)
- Jingyu Cao
- Department of Oncology, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, China
| | - Xinyi Ma
- The First Clinical Medical College, Southern Medical University, Guangzhou, China
| | - Guijuan Zhang
- School of Nursing of Jinan University, Guangzhou, China
| | - Shouyi Hong
- College of Traditional Chinese Medicine, Institute of Integrated Traditional Chinese and Western Medicine, Jinan University, Guangzhou, China
| | - Ruirui Ma
- College of Traditional Chinese Medicine, Institute of Integrated Traditional Chinese and Western Medicine, Jinan University, Guangzhou, China
| | - Yanqiu Wang
- College of Traditional Chinese Medicine, Institute of Integrated Traditional Chinese and Western Medicine, Jinan University, Guangzhou, China
| | - Xianxin Yan
- College of Traditional Chinese Medicine, Institute of Integrated Traditional Chinese and Western Medicine, Jinan University, Guangzhou, China
| | - Min Ma
- College of Traditional Chinese Medicine, Institute of Integrated Traditional Chinese and Western Medicine, Jinan University, Guangzhou, China
| |
Collapse
|
4
|
Feuerer L, Lamm S, Henz I, Kappelmann-Fenzl M, Haferkamp S, Meierjohann S, Hellerbrand C, Kuphal S, Bosserhoff AK. Role of melanoma inhibitory activity in melanocyte senescence. Pigment Cell Melanoma Res 2019; 32:777-791. [PMID: 31172672 DOI: 10.1111/pcmr.12801] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Revised: 05/16/2019] [Accepted: 05/26/2019] [Indexed: 01/10/2023]
Abstract
The protein melanoma inhibitory activity (MIA) is known to be expressed in melanoma and to support melanoma progression. Interestingly, previous studies also observed the expression of MIA in nevi. Concentrating on these findings, we revealed that MIA expression is correlated with a senescent state in melanocytes. Induction of replicative or oncogene-induced senescence resulted in increased MIA expression in vitro. Notably, MIA knockdown in senescent melanocytes reduced the percentage of senescence-associated beta-Gal-positive cells and enhanced proliferation. Using the melanoma mouse model Tg(Grm1), MIA-deficient mice supported the impact of MIA on senescence by showing a significantly earlier tumor onset compared to controls. In melanocytes, MIA knockdown led to a downregulation of the cell cycle inhibitor p21 in vitro and in vivo. In contrast, after induction of hTERT in human melanoma cells, p21 regulation by MIA was lost. In summary, our data show for the first time that MIA is a regulator of cellular senescence in human and murine melanocytes.
Collapse
Affiliation(s)
- Lena Feuerer
- Emil Fischer Center, Institute of Biochemistry, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Susanne Lamm
- Emil Fischer Center, Institute of Biochemistry, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Ingmar Henz
- Emil Fischer Center, Institute of Biochemistry, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Melanie Kappelmann-Fenzl
- Emil Fischer Center, Institute of Biochemistry, University of Erlangen-Nürnberg, Erlangen, Germany.,Deggendorf Institute of Technology, Deggendorf, Germany
| | - Sebastian Haferkamp
- Department of Dermatology, University Hospital Regensburg, Regensburg, Germany
| | | | - Claus Hellerbrand
- Emil Fischer Center, Institute of Biochemistry, University of Erlangen-Nürnberg, Erlangen, Germany.,Comprehensive Cancer Center (CCC) Erlangen-EMN, Erlangen, Germany
| | - Silke Kuphal
- Emil Fischer Center, Institute of Biochemistry, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Anja Katrin Bosserhoff
- Emil Fischer Center, Institute of Biochemistry, University of Erlangen-Nürnberg, Erlangen, Germany.,Comprehensive Cancer Center (CCC) Erlangen-EMN, Erlangen, Germany
| |
Collapse
|
5
|
Melanom. MEDIKAMENTÖSE TUMORTHERAPIE IN DER DERMATO-ONKOLOGIE 2019. [PMCID: PMC7121576 DOI: 10.1007/978-3-662-58012-7_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Das Melanom ist der gefährlichste Hautkrebs mit der höchsten Sterblichkeitsrate, der schon bei jungen Menschen auftreten kann und seit Jahrzehnten steigende Inzidenz verzeichnet (Jemal et al. 2007; Little et al. 2012). Jährlich erkranken weltweit etwa 137.000 Menschen am Melanom und 37.000 versterben an der Erkrankung (Boyle et al. 2004). Die Inzidenz liegt weltweit jährlich bei 2,3–2,6/100.000 Einwohner (Pisani et al. 2002). In Deutschland beträgt die Inzidenz 19,2/100.000 Einwohner und es verstarben 2711 Betroffene im Jahre 2010 (Statistisches Bundesamt).
Collapse
|
6
|
Sasahira T, Bosserhoff AK, Kirita T. The importance of melanoma inhibitory activity gene family in the tumor progression of oral cancer. Pathol Int 2018; 68:278-286. [PMID: 29655307 DOI: 10.1111/pin.12672] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Accepted: 03/14/2018] [Indexed: 01/14/2023]
Abstract
Oral squamous cell carcinoma has a high potential for locoregional invasion and nodal metastasis. Consequently, early detection of such malignancies is of immense importance. The melanoma inhibitory activity (MIA) gene family comprises MIA, MIA2, transport and Golgi organization protein 1 (TANGO), and otoraplin (OTOR). These members of the MIA gene family have a highly conserved Src homology 3 (SH3)-like structure. Although the molecules of this family share 34-45% amino acid homology and 47-59% cDNA sequence homology, those members, excluding OTOR, play different tumor-associated functions. MIA has a pivotal role in the progression and metastasis of melanoma; MIA2 and TANGO have been suggested to possess tumor-suppressive functions; and OTOR is uniquely expressed in cochlea of the inner ear. Therefore, the definite functions of the MIA gene family in cancer cells remain unclear. Since the members of the MIA gene family are secreted proteins, these molecules might be useful tumor markers that can be detected in the body fluids, including serum and saliva. In this review, we described the molecular biological functions of the MIA gene family in oral cancer.
Collapse
Affiliation(s)
- Tomonori Sasahira
- Department of Molecular Pathology, Nara Medical University, Kashihara, Japan
| | - Anja Katrin Bosserhoff
- Institute for Biochemistry, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Tadaaki Kirita
- Department of Oral and Maxillofacial Surgery, Nara Medical University, Kashihara, Japan
| |
Collapse
|
7
|
Eisenstein A, Gonzalez EC, Raghunathan R, Xu X, Wu M, McLean EO, McGee J, Ryu B, Alani RM. Emerging Biomarkers in Cutaneous Melanoma. Mol Diagn Ther 2018; 22:203-218. [PMID: 29411301 DOI: 10.1007/s40291-018-0318-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Earlier identification of aggressive melanoma remains a goal in the field of melanoma research. With new targeted and immune therapies that have revolutionized the care of patients with melanoma, the ability to predict progression and monitor or predict response to therapy has become the new focus of research into biomarkers in melanoma. In this review, promising biomarkers are highlighted. These biomarkers have been used to diagnose melanoma as well as predict progression to advanced disease and response to therapy. The biomarkers take various forms, including protein expression at the level of tissue, genetic mutations of cancer cells, and detection of circulating DNA. First, a brief description is provided about the conventional tissue markers used to stage melanoma, including tumor depth. Next, protein biomarkers, which provide both diagnostic and prognostic information, are described. This is followed by a discussion of important genetic mutations, microRNA, and epigenetic modifications that can provide therapeutic and prognostic material. Finally, emerging serologic biomarkers are reviewed, including circulating melanoma cells and exosomes. Overall the goal is to identify biomarkers that aid in the earlier identification and improved treatment of aggressive melanoma.
Collapse
Affiliation(s)
- Anna Eisenstein
- Department of Dermatology, Boston University School of Medicine, 609 Albany Street, Boston, MA, 02118, USA
| | - Estela Chen Gonzalez
- Department of Dermatology, Boston University School of Medicine, 609 Albany Street, Boston, MA, 02118, USA
| | - Rekha Raghunathan
- Department of Dermatology, Boston University School of Medicine, 609 Albany Street, Boston, MA, 02118, USA
| | - Xixi Xu
- Department of Dermatology, Boston University School of Medicine, 609 Albany Street, Boston, MA, 02118, USA
| | - Muzhou Wu
- Department of Dermatology, Boston University School of Medicine, 609 Albany Street, Boston, MA, 02118, USA
| | - Emily O McLean
- Department of Dermatology, Boston University School of Medicine, 609 Albany Street, Boston, MA, 02118, USA
| | - Jean McGee
- Department of Dermatology, Boston University School of Medicine, 609 Albany Street, Boston, MA, 02118, USA
| | - Byungwoo Ryu
- Department of Dermatology, Boston University School of Medicine, 609 Albany Street, Boston, MA, 02118, USA.
| | - Rhoda M Alani
- Department of Dermatology, Boston University School of Medicine, 609 Albany Street, Boston, MA, 02118, USA.
| |
Collapse
|
8
|
Sasahira T, Nishiguchi Y, Fujiwara R, Kurihara M, Kirita T, Bosserhoff AK, Kuniyasu H. Storkhead box 2 and melanoma inhibitory activity promote oral squamous cell carcinoma progression. Oncotarget 2018; 7:26751-64. [PMID: 27050375 PMCID: PMC5042012 DOI: 10.18632/oncotarget.8495] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 03/10/2016] [Indexed: 11/25/2022] Open
Abstract
Background Storkhead box protein 2 (STOX2) is a transcriptional factor associated with pre-eclampsia with fetal growth restriction. We recently reported that melanoma inhibitory activity (MIA) promotes oral squamous cell carcinoma (OSCC) progression. However, the relationship between STOX2 and MIA remains unknown in malignancies. Methods We used immunohistochemistry and PCR to investigate MIA and STOX2 expression in OSCC. We also performed functional analysis in human OSCC cells. Results MIA and STOX2 mRNA levels were higher in OSCCs than in normal oral epithelial cells, and upregulation of STOX2 was significantly correlated with overexpression of MIA. Immunostaining for STOX2 was associated with nodal metastasis (P = 0.0002) and MIA expression (P < 0.0001). Furthermore, MIA expression (P = 0.0035) and STOX2 expression (P = 0.0061) were associated with poor outcome in OSCCs. In vitro analysis using OSCC cells revealed that MIA increased expression of STOX2 by paracrine manner. Moreover, STOX2 accelerated OSCC cell growth, invasion, suppressed apoptosis, and enhanced resistance to paclitaxel, cisplatin, and 5-FU. Conclusions Our results suggest that MIA-STOX2 signaling may be a useful diagnostic and therapeutic target in OSCCs.
Collapse
Affiliation(s)
- Tomonori Sasahira
- Department of Molecular Pathology, Nara Medical University, Kashihara, Japan
| | - Yukiko Nishiguchi
- Department of Molecular Pathology, Nara Medical University, Kashihara, Japan
| | - Rina Fujiwara
- Department of Molecular Pathology, Nara Medical University, Kashihara, Japan
| | - Miyako Kurihara
- Department of Molecular Pathology, Nara Medical University, Kashihara, Japan.,Department of Oral and Maxillofacial Surgery, Nara Medical University, Kashihara, Japan
| | - Tadaaki Kirita
- Department of Oral and Maxillofacial Surgery, Nara Medical University, Kashihara, Japan
| | - Anja Katrin Bosserhoff
- Institute for Biochemistry, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Hiroki Kuniyasu
- Department of Molecular Pathology, Nara Medical University, Kashihara, Japan
| |
Collapse
|
9
|
Li B, He X, Zhao Y, Bai D, Shiraigo W, Zhao Q, Manglai D. Regulatory pathway analysis of coat color genes in Mongolian horses. Hereditas 2017; 155:13. [PMID: 28974924 PMCID: PMC5622463 DOI: 10.1186/s41065-017-0048-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 09/18/2017] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Studies on the molecular genetics of horse skin pigmentation have typically focused on very few genes and proteins. In this study, we used Illumina sequencing to determine the global gene expression profiles in horses with white-colored coats and those with black-colored coats, with the goal of identifying novel genes that could regulate horse coat color. RESULTS Genes encoding ribosomal-associated proteins were highly expressed in horse skin. We found a total of 231 unigenes that were differentially expressed between horses with white coats and horses with black coats; 119 were down-regulated, and 112 were up-regulated. Many of the up-regulated genes in black horses, such as genes related to tyrosine metabolism, may directly regulate dark coat color. Keratin genes, MIA family genes, fatty acid-related genes, and melanoma-associated genes were also differentially regulated, which suggests that they may play important roles in coat color formation. CONCLUSIONS These findings show that the transcription profiles from white and black horse skin provide useful information to understand the genetics underlying the control of skin melanin synthesis in horses, which may enhance our knowledge of human skin diseases, such as melanoma and albinism.
Collapse
Affiliation(s)
- Bei Li
- College of Animal Science, Inner Mongolia Agricultural University, Huhhot, 010018 People’s Republic of China
| | - Xiaolong He
- Inner Mongolia Academy of Agricultural and Animal Husbandry Sciences, Huhhot, 010031 People’s Republic of China
| | - Yiping Zhao
- College of Animal Science, Inner Mongolia Agricultural University, Huhhot, 010018 People’s Republic of China
| | - Dongyi Bai
- College of Animal Science, Inner Mongolia Agricultural University, Huhhot, 010018 People’s Republic of China
| | - Wunierfu Shiraigo
- College of Animal Science, Inner Mongolia Agricultural University, Huhhot, 010018 People’s Republic of China
| | - Qinan Zhao
- College of Animal Science, Inner Mongolia Agricultural University, Huhhot, 010018 People’s Republic of China
| | - Dugarjaviin Manglai
- College of Animal Science, Inner Mongolia Agricultural University, Huhhot, 010018 People’s Republic of China
| |
Collapse
|
10
|
Alegre E, Zubiri L, Perez-Gracia JL, González-Cao M, Soria L, Martín-Algarra S, González A. Circulating melanoma exosomes as diagnostic and prognosis biomarkers. Clin Chim Acta 2015; 454:28-32. [PMID: 26724367 DOI: 10.1016/j.cca.2015.12.031] [Citation(s) in RCA: 129] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 12/11/2015] [Accepted: 12/21/2015] [Indexed: 02/07/2023]
Abstract
BACKGROUND Malignant melanoma is an aggressive cancer with an increasing incidence. Exosomes are actively secreted microvesicles, whose characteristics reflect those of the cell they are originated in. The aim of this study was to identify and evaluate the presence of the melanoma biomarkers MIA, S100B and tyrosinase-related protein 2 (TYRP2) in exosomes and their potential clinical utility. METHODS Serum samples were obtained from stage IV melanoma patients, melanoma-free patients and healthy controls. Exosomes were precipitated and TYRP2, MIA and S100B concentrations were quantified in serum, exosomes, and exosome-free serum. RESULTS Both MIA and S100B were detected in exosomes and correlated significantly with serum concentrations (S100B: r=0.968; MIA: r=0.799; p<0.001). MIA and S100B concentrations in exosomes were significantly higher in melanoma patients than in healthy controls and disease-free patients. However, TYRP2 concentrations in exosomes did not differ between these three groups. ROC curves analysis rendered AUCs for MIA of 0.883 (p<0.01) and of 0.840 for S100B (p<0.01). Patients with exosome MIA concentration higher than 2.5 μg/L showed shorter median survival related to those with lower level (4 versus 11 months; p<0.05). CONCLUSIONS MIA and S100B can be detected in exosomes from melanoma patients and their quantification presents diagnostic and prognostic utility.
Collapse
Affiliation(s)
| | - Leyre Zubiri
- Department of Medical Oncology, University Clinic of Navarra, Spain
| | | | - María González-Cao
- Translational Cancer Research Unit, Instituto Oncológico Dr Rosell, Quirón Dexeus University Hospital, Barcelona, Spain
| | - Lourdes Soria
- Department of Medical Oncology, University Clinic of Navarra, Spain
| | | | - Alvaro González
- Laboratory of Biochemistry, University Clinic of Navarra, Spain.
| |
Collapse
|
11
|
Odashiro M, Hans Filho G, Pereira PR, Castro ARCM, Stief AC, Pontes ERJC, Odashiro AN. Melanoma inhibitory activity in Brazilian patients with cutaneous melanoma. An Bras Dermatol 2015; 90:327-32. [PMID: 26131861 PMCID: PMC4516101 DOI: 10.1590/abd1806-4841.20153248] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 07/31/2014] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Melanoma inhibitory activity is a protein secreted by melanoma cells and has been used as a tumor marker. Increased Melanoma inhibitory activity serum levels are related to metastatic disease or tumor recurrence. Currently there are no studies on Melanoma inhibitory activity and cutaneous melanoma involving Brazilian patients. OBJECTIVE To evaluate the performance and feasibility of measuring Melanoma inhibitory activity levels in Brazilian patients with cutaneous melanoma. METHODS Blood was obtained from ten patients with proved metastatic cutaneous melanoma (Group 1), 15 patients resected for cutaneous melanoma without metastasis (Group 2) and 5 healthy donors (Group 3). Melanoma inhibitory activity was measured using a commercially available ELISA kit. RESULTS There was a statistically significant difference of Melanoma inhibitory activity levels between patients with and without metastasis (p=0.002), and between patients with metastasis and healthy donors (p=0.002). There was no difference between patients without metastasis and healthy donors (p=0.443). CONCLUSION Melanoma inhibitory activity is a tumor marker for cutaneous melanoma and the Melanoma inhibitory activity-ELISA test can be easily performed. Patients with metastasis have increased Melanoma inhibitory activity serum levels when compared to patients without metastasis and healthy donors.
Collapse
Affiliation(s)
- Macanori Odashiro
- Universidade Federal do Mato Grosso do Sul, Campo Grande, MS, Brazil
| | - Gunter Hans Filho
- Universidade Federal do Mato Grosso do Sul, Campo Grande, MS, Brazil
| | | | | | | | | | | |
Collapse
|
12
|
Sandru A, Voinea S, Panaitescu E, Bolovan M, Stanciu A, Cinca S, Blidaru A. Clinical value of Melanoma Inhibitory Activity in stratifying malignant melanoma patients. Lab Invest 2015. [PMCID: PMC4315275 DOI: 10.1186/1479-5876-13-s1-p10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
13
|
Riechers A, Bosserhoff AK. Melanoma inhibitory activity in melanoma diagnostics and therapy - a small protein is looming large. Exp Dermatol 2014; 23:12-4. [PMID: 24372647 DOI: 10.1111/exd.12281] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/01/2013] [Indexed: 12/15/2022]
Abstract
Malignant melanoma is a highly aggressive cancer with a very poor prognosis after the onset of metastasis. We have previously demonstrated that the protein melanoma inhibitory activity (MIA) is involved in the metastasis of and immunosuppression in malignant melanoma. Recently, we further established MIA as a therapeutic target to inhibit metastatic spread in malignant melanoma. We could show that an inhibition of MIA by a synthetic peptide decreased both the number of metastases as well as immunosuppression in a murine model of malignant melanoma. To control recurrence after surgical resection of a primary lesion, it is paramount to have diagnostic tools available that can detect a relapse due to the strong metastatic potential of melanoma. This follow-up is maintained with periodic re-examinations. Due to high cost and the associated radiation exposure, radiology examinations are avoided if possible. The analysis of prognostic markers in patient serum is therefore attractive. In this review, we focus on the quantitative analysis of the MIA protein as a prognostic tool because it has proven to be a useful serum marker for documenting disease progression of malignant melanoma. The MIA quantification assay itself is readily performed using an ELISA kit and common laboratory equipment. Because analysing MIA serum levels in combination with other established markers such as S100B improves their prognostic value, we feel that the quantification of MIA in the serum, among other markers, should be performed as a general standard of care in patients at risk of developing metastatic melanoma.
Collapse
|
14
|
Aladowicz E, Ferro L, Vitali GC, Venditti E, Fornasari L, Lanfrancone L. Molecular networks in melanoma invasion and metastasis. Future Oncol 2013; 9:713-26. [PMID: 23647299 DOI: 10.2217/fon.13.9] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Metastatic melanoma accounts for approximately 80% of skin cancer-related deaths. Up to now there has been no effective treatment for stage IV melanoma patients due to the complexity and dissemination potential of this disease. Melanomas are heterogeneous tumors in which conventional therapies fail to improve overall survival. Targeted therapies are being developed, but the final outcome can be hampered by the incomplete knowledge of the process of melanoma progression. Even if the intracellular pathways are similar, the interaction of the cells with the surrounding environment should be taken into consideration. This article seeks to highlight some of the advances in the understanding of the molecular mechanisms underlying melanoma dissemination.
Collapse
Affiliation(s)
- Ewa Aladowicz
- Department of Experimental Oncology, European Institute of Oncology, Via Adamello 16, Milan, Italy
| | | | | | | | | | | |
Collapse
|
15
|
Ivanov SV, Panaccione A, Nonaka D, Prasad ML, Boyd KL, Brown B, Guo Y, Sewell A, Yarbrough WG. Diagnostic SOX10 gene signatures in salivary adenoid cystic and breast basal-like carcinomas. Br J Cancer 2013; 109:444-51. [PMID: 23799842 PMCID: PMC3721393 DOI: 10.1038/bjc.2013.326] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2013] [Revised: 06/01/2013] [Accepted: 06/04/2013] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Salivary adenoid cystic carcinoma (ACC) is an insidious slow-growing cancer with the propensity to recur and metastasise to distant sites. Basal-like breast carcinoma (BBC) is a molecular subtype that constitutes 15-20% of breast cancers, shares histological similarities and basal cell markers with ACC, lacks expression of ER (oestrogen receptor), PR (progesterone receptor), and HER2 (human epidermal growth factor receptor 2), and, similar to ACC, metastasises predominantly to the lung and brain. Both cancers lack targeted therapies owing to poor understanding of their molecular drivers. METHODS Gene expression profiling, immunohistochemical staining, western blot, RT-PCR, and in silico analysis of massive cancer data sets were used to identify novel markers and potential therapeutic targets for ACC and BBC. For the detection and comparison of gene signatures, we performed co-expression analysis using a recently developed web-based multi-experiment matrix tool for visualisation and rank aggregation. RESULTS In ACC and BBC we identified characteristic and overlapping SOX10 gene signatures that contained a large set of novel potential molecular markers. SOX10 was validated as a sensitive diagnostic marker for both cancers and its expression was linked to normal and malignant myoepithelial/basal cells. In ACC, BBC, and melanoma (MEL), SOX10 expression strongly co-segregated with the expression of ROPN1B, GPM6B, COL9A3, and MIA. In ACC and breast cancers, SOX10 expression negatively correlated with FOXA1, a cell identity marker and major regulator of the luminal breast subtype. Diagnostic significance of several conserved elements of the SOX10 signature (MIA, TRIM2, ROPN1, and ROPN1B) was validated on BBC cell lines. CONCLUSION SOX10 expression in ACC and BBC appears to be a part of a highly coordinated transcriptional programme characteristic for cancers with basal/myoepithelial features. Comparison between ACC/BBC and other cancers, such as neuroblastomaand MEL, reveals potential molecular markers specific for these cancers that are likely linked to their cell identity. SOX10 as a novel diagnostic marker for ACC and BBC provides important molecular insight into their molecular aetiology and cell origin. Given that SOX10 was recently described as a principal driver of MEL, identification of conserved elements of the SOX10 signatures may help in better understanding of SOX10-related signalling and development of novel diagnostic and therapeutic tools.
Collapse
Affiliation(s)
- S V Ivanov
- Section of Otolaryngology, Department of Surgery, Yale School of Medicine, 800 Howard Avenue, New Haven, CT 06519-1369, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Wang G, Lunardi A, Zhang J, Chen Z, Ala U, Webster KA, Tay Y, Gonzalez-Billalabeitia E, Egia A, Shaffer DR, Carver B, Liu XS, Taulli R, Kuo WP, Nardella C, Signoretti S, Cordon-Cardo C, Gerald WL, Pandolfi PP. Zbtb7a suppresses prostate cancer through repression of a Sox9-dependent pathway for cellular senescence bypass and tumor invasion. Nat Genet 2013; 45:739-746. [PMID: 23727861 PMCID: PMC4036521 DOI: 10.1038/ng.2654] [Citation(s) in RCA: 129] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Accepted: 05/02/2013] [Indexed: 12/12/2022]
Abstract
Lrf has been previously described as a powerful proto-oncogene. Here we surprisingly demonstrate that Lrf plays a critical oncosuppressive role in the prostate. Prostate specific inactivation of Lrf leads to a dramatic acceleration of Pten-loss-driven prostate tumorigenesis through a bypass of Pten-loss-induced senescence (PICS). We show that LRF physically interacts with and functionally antagonizes SOX9 transcriptional activity on key target genes such as MIA, which is involved in tumor cell invasion, and H19, a long non-coding RNA precursor for an Rb-targeting miRNA. Inactivation of Lrf in vivo leads to Rb down-regulation, PICS bypass and invasive prostate cancer. Importantly, we found that LRF is genetically lost, as well as down-regulated at both the mRNA and protein levels in a subset of human advanced prostate cancers. Thus, we identify LRF as a context-dependent cancer gene that can act as an oncogene in some contexts but also displays oncosuppressive-like activity in Pten−/− tumors.
Collapse
Affiliation(s)
- Guocan Wang
- Cancer Genetics Program, Beth Israel Deaconess Cancer Center, Departments of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA.,BCMB Program, Weill Graduate School of Medical Sciences, Cornell University, New York, New York 10021.,Cancer Biology and Genetics Program, Sloan-Kettering Institute, 1275 York Avenue, New York, New York 10021, USA.,Department of Pathology, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, New York 10021, USA
| | - Andrea Lunardi
- Cancer Genetics Program, Beth Israel Deaconess Cancer Center, Departments of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA
| | - Jiangwen Zhang
- FAS Center for Systems Biology, Harvard University, Cambridge, Massachusetts 02138, USA
| | - Zhenbang Chen
- Cancer Biology and Genetics Program, Sloan-Kettering Institute, 1275 York Avenue, New York, New York 10021, USA.,Department of Pathology, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, New York 10021, USA
| | - Ugo Ala
- Cancer Genetics Program, Beth Israel Deaconess Cancer Center, Departments of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA
| | - Kaitlyn A Webster
- Cancer Genetics Program, Beth Israel Deaconess Cancer Center, Departments of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA
| | - Yvonne Tay
- Cancer Genetics Program, Beth Israel Deaconess Cancer Center, Departments of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA
| | - Enrique Gonzalez-Billalabeitia
- Cancer Genetics Program, Beth Israel Deaconess Cancer Center, Departments of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA
| | - Ainara Egia
- Cancer Genetics Program, Beth Israel Deaconess Cancer Center, Departments of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA
| | - David R Shaffer
- Cancer Biology and Genetics Program, Sloan-Kettering Institute, 1275 York Avenue, New York, New York 10021, USA.,Department of Pathology, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, New York 10021, USA
| | - Brett Carver
- Human Oncology and Pathogenesis Program, Department of Surgery, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, New York 10021, USA
| | - Xue-Song Liu
- Cancer Genetics Program, Beth Israel Deaconess Cancer Center, Departments of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA
| | - Riccardo Taulli
- Cancer Genetics Program, Beth Israel Deaconess Cancer Center, Departments of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA
| | - Winston Patrick Kuo
- Department of Developmental Biology, Harvard School Of Dental Medicine, Boston, MA 02115, USA
| | - Caterina Nardella
- Cancer Genetics Program, Beth Israel Deaconess Cancer Center, Departments of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA.,Cancer Biology and Genetics Program, Sloan-Kettering Institute, 1275 York Avenue, New York, New York 10021, USA.,Department of Pathology, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, New York 10021, USA.,Preclinical Murine Pharmacogenetics Facility, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Sabina Signoretti
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA. MA.,Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Carlos Cordon-Cardo
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, New York 10021, USA
| | - William L Gerald
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, New York 10021, USA
| | - Pier Paolo Pandolfi
- Cancer Genetics Program, Beth Israel Deaconess Cancer Center, Departments of Medicine and Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA.,BCMB Program, Weill Graduate School of Medical Sciences, Cornell University, New York, New York 10021.,Cancer Biology and Genetics Program, Sloan-Kettering Institute, 1275 York Avenue, New York, New York 10021, USA.,Department of Pathology, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, New York 10021, USA
| |
Collapse
|
17
|
Tuan TF, Chung CT, Tsou HH, Chen FW, Lin HL, Lai YK, Lee WS, Chao YS, Hwang LL, Chen CT. Putative tumor metastasis-associated genes in human gastric cancer. Int J Oncol 2012; 41:1068-84. [PMID: 22664961 DOI: 10.3892/ijo.2012.1502] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2012] [Accepted: 05/11/2012] [Indexed: 01/06/2023] Open
Abstract
Gastric cancer is one of the leading causes of cancer mortality and its malignancy, resulting from disseminated cancer cells of diffuse type, is clinically manifested as metastases to the liver and peritoneum. The aim of the present study was to identify putative tumor metastasis-associated genes in human gastric cancer cells of diffuse type. An MKN45 cell line constitutively expressing green fluorescent protein (MKN45-GFP) was established and selected using the Transwell® system for invasive sublines MKN45-GFP-4, MKN45-GFP-10 and MKN45-GFP-12. MKN45-GFP-10 and MKN45-GFP-12 are highly invasive compared to the others. The mRNA levels were measured with cDNA microarrays and correlated with their invasion abilities in these sublines. Many of the genes identified with a positive or negative correlation are associated with angiogenesis, cell cycle, cytoskeleton and cell motility, protease and cell adhesion, as well as cellular signal transduction. In particular, novel genes without known functions were also noted. RT-PCR and western blot analyses were applied to verify the expression of selective genes. Following orthotopical intraperitoneal implantation, MKN45-GFP-12 demonstrated significantly higher in vivo tumor malignancies than parental MKN45-GFP in ascites induction and liver -invasion in mice. We have identified putative gastric tumor metastasis-associated, as well as novel genes. These genes and their protein products are to be further explored for their functional roles associated with tumor metastasis. The molecular profiles of these identified genes, gene transcripts and proteins in the patient specimens are likely to be useful biomarkers for diagnostic, therapeutic and/or prognostics. Most importantly, they may be used as molecular targets for the discovery of antitumor drugs against human gastric cancer metastasis.
Collapse
Affiliation(s)
- Tsung-Fan Tuan
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan, R.O.C
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Schmidt J, Riechers A, Stoll R, Amann T, Fink F, Spruss T, Gronwald W, König B, Hellerbrand C, Bosserhoff AK. Targeting melanoma metastasis and immunosuppression with a new mode of melanoma inhibitory activity (MIA) protein inhibition. PLoS One 2012; 7:e37941. [PMID: 22666418 PMCID: PMC3362532 DOI: 10.1371/journal.pone.0037941] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2012] [Accepted: 04/26/2012] [Indexed: 11/18/2022] Open
Abstract
Melanoma is the most aggressive form of skin cancer, with fast progression and early dissemination mediated by the melanoma inhibitory activity (MIA) protein. Here, we discovered that dimerization of MIA is required for functional activity through mutagenesis of MIA which showed the correlation between dimerization and functional activity. We subsequently identified the dodecapeptide AR71, which prevents MIA dimerization and thereby acts as a MIA inhibitor. Two-dimensional nuclear magnetic resonance (NMR) spectroscopy demonstrated the binding of AR71 to the MIA dimerization domain, in agreement with in vitro and in vivo data revealing reduced cell migration, reduced formation of metastases and increased immune response after AR71 treatment. We believe AR71 is a lead structure for MIA inhibitors. More generally, inhibiting MIA dimerization is a novel therapeutic concept in melanoma therapy.
Collapse
Affiliation(s)
- Jennifer Schmidt
- Institute of Pathology, University of Regensburg, Regensburg, Germany
| | - Alexander Riechers
- Institute of Organic Chemistry, University of Regensburg, Regensburg, Germany
| | - Raphael Stoll
- Faculty of Chemistry and Biochemistry, Biomolecular NMR, Ruhr University of Bochum, Bochum, Germany
| | - Thomas Amann
- Department of Internal Medicine I, University Hospital Regensburg, Regensburg, Germany
| | - Florian Fink
- Institute of Functional Genomics, University of Regensburg, Regensburg, Germany
| | - Thilo Spruss
- Institute of Pharmacy, University of Regensburg, Regensburg, Germany
| | - Wolfram Gronwald
- Institute of Functional Genomics, University of Regensburg, Regensburg, Germany
| | - Burkhard König
- Institute of Organic Chemistry, University of Regensburg, Regensburg, Germany
| | - Claus Hellerbrand
- Department of Internal Medicine I, University Hospital Regensburg, Regensburg, Germany
| | | |
Collapse
|
19
|
Current World Literature. Curr Opin Support Palliat Care 2010; 4:293-304. [DOI: 10.1097/spc.0b013e328340e983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
20
|
Heller A, Zörnig I, Müller T, Giorgadze K, Frei C, Giese T, Bergmann F, Schmidt J, Werner J, Buchler MW, Jaeger D, Giese NA. Immunogenicity of SEREX-identified antigens and disease outcome in pancreatic cancer. Cancer Immunol Immunother 2010; 59:1389-400. [PMID: 20514540 PMCID: PMC11029919 DOI: 10.1007/s00262-010-0870-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2010] [Accepted: 05/15/2010] [Indexed: 01/06/2023]
Abstract
Despite spontaneous or vaccination-induced immune responses, pancreatic cancer remains one of the most deadly immunotherapy-resistant malignancies. We sought to comprehend the spectrum of pancreatic tumor-associated antigens (pTAAs) and to assess the clinical relevance of their immunogenicity. An autologous SEREX-based screening of a cDNA library constructed from a pancreatic T3N0M0/GIII specimen belonging to a long-term survivor (36 months) revealed 18 immunogenic pTAA. RT-PCR analysis displayed broad distribution of the identified antigens among normal human tissues. PNLIPRP2 and MIA demonstrated the most distinct pancreatic cancer-specific patterns. ELISA-based screening of sera for corresponding autoantibodies revealed that although significantly increased, the immunogenicity of these molecules was not a common feature in pancreatic cancer. QRT-PCR and immunohistochemistry characterized PNLIPRP2 as a robust acinar cell-specific marker whose decreased expression mirrored the disappearance of parenchyma in the diseased organ, but was not related to the presence of PNLIPRP2 autoantibodies. Analyses of MIA-known to be preferentially expressed in malignant cells-surprisingly revealed an inverse correlation between intratumoral gene expression and the emergence of autoantibodies. MIA(high) patients were autoantibody-negative and had shorter median survival when compared with autoantibody-positive MIA(low) patients (12 vs. 34 months). The observed pTAA spectrum comprised molecules associated with acinar, stromal and malignant structures, thus presenting novel targets for tumor cell-specific therapies as well as for approaches based on the bystander effects. Applying the concept of cancer immunoediting to interpret relationships between gene expression, antitumor immune responses, and clinical outcome might better discriminate between past and ongoing immune responses, consequently enabling prognostic stratification of patients and individual adjustment of immunotherapy.
Collapse
Affiliation(s)
- A. Heller
- Department of Surgery, University Hospital Heidelberg, INF 116, 69120 Heidelberg, Germany
| | - I. Zörnig
- Medical Oncology, National Centre of Tumor Diseases (NCT), University Hospital Heidelberg, INF 350, 69120 Heidelberg, Germany
| | - T. Müller
- Medical Oncology, National Centre of Tumor Diseases (NCT), University Hospital Heidelberg, INF 350, 69120 Heidelberg, Germany
| | - K. Giorgadze
- Department of Surgery, University Hospital Heidelberg, INF 116, 69120 Heidelberg, Germany
| | - C. Frei
- Medical Oncology, National Centre of Tumor Diseases (NCT), University Hospital Heidelberg, INF 350, 69120 Heidelberg, Germany
| | - T. Giese
- Institute of Immunology, University Hospital Heidelberg, INF 305, 69120 Heidelberg, Germany
| | - F. Bergmann
- Institute of Pathology, University Hospital Heidelberg, INF 220, 69120 Heidelberg, Germany
| | - J. Schmidt
- Department of Surgery, University Hospital Heidelberg, INF 116, 69120 Heidelberg, Germany
| | - J. Werner
- Department of Surgery, University Hospital Heidelberg, INF 116, 69120 Heidelberg, Germany
| | - M. W. Buchler
- Department of Surgery, University Hospital Heidelberg, INF 116, 69120 Heidelberg, Germany
| | - D. Jaeger
- Medical Oncology, National Centre of Tumor Diseases (NCT), University Hospital Heidelberg, INF 350, 69120 Heidelberg, Germany
| | - N. A. Giese
- Department of Surgery, University Hospital Heidelberg, INF 116, 69120 Heidelberg, Germany
| |
Collapse
|
21
|
Migration-associated secretion of melanoma inhibitory activity at the cell rear is supported by KCa3.1 potassium channels. Cell Res 2010; 20:1224-38. [DOI: 10.1038/cr.2010.121] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
|
22
|
Heterogeneous transition metal-based fluorescence polarization (HTFP) assay for probing protein interactions. Biotechniques 2010; 47:837-44. [PMID: 19852767 DOI: 10.2144/000113223] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Analyses of protein interactions are fundamental for the investigation of molecular mechanisms responsible for cellular processes and diseases, as well as for drug discovery in the pharmaceutical industry. The present study details the development of a fluorescence polarization assay using melanoma inhibitory activity (MIA) protein-binding compounds and studies of the binding properties of this protein. Since they are dependent on the the lifetime of the fluorescent label, currently available fluorescence polarization assays can only determine interactions with either high- or low-molecular weight interaction partners. Our new approach eliminates this limitation by immobilizing a known binding partner of MIA protein to a well plate and by labeling the target protein using luminescent transition metal labels such as Ru(bpy)3 for binding studies with both high- and low-molecular weight interaction partners. Due to the use of a functionalized surface, we termed our concept heterogeneous transition metal-based fluorescence polarization (HTFP) assay. The assay's independence from the molecular weight of potential binding partners should make the technique amenable to investigations on subjects as diverse as multimerization, interactions with pharmacophores, or binding affinity determination.
Collapse
|