1
|
Juza A, Kołodziej-Spirodek L, Gutkowski K, Partyka M, Dąbrowski M. Distinguishing exocrine pancreas disease-associated diabetes from type 2 diabetes based on anthropometric and metabolic parameters. World J Diabetes 2025; 16:95102. [PMID: 39959260 PMCID: PMC11718472 DOI: 10.4239/wjd.v16.i2.95102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 08/28/2024] [Accepted: 10/29/2024] [Indexed: 12/30/2024] Open
Abstract
BACKGROUND Adult-onset diabetes is most often considered to be type 2 diabetes. However, other types of diabetes can develop in adults, including exocrine pancreas disease-associated diabetes, also called type 3c diabetes. Differential diagnosis between these types of diabetes still remains a diagnostic challenge. AIM To define anthropometric and laboratory markers that will allow for early diagnosis of pancreatic disease-associated diabetes. METHODS The study group included 44 patients with pancreatogenic diabetes (26 with pancreatic cancer and 18 with chronic pancreatitis), while the control group consisted of 35 patients with type 2 diabetes. We analyzed several parameters, including sex, age, body mass index (BMI), fasting plasma glucose, fasting C-peptide and insulin with homeostasis model assessment of insulin resistance (HOMA-IR) index calculation, adrenomedullin, adiponectin and creatinine levels with epidermal growth factor receptor (eGFR) calculation. We also developed an equation, termed type 3c diabetes index, which utilized BMI, fasting insulin and adrenomedullin levels, and eGFR to better identify patients with type 3c diabetes. RESULTS Compared to patients with type 2 diabetes, patients with pancreatogenic diabetes had significantly lower BMI (25.11 ± 4.87 kg/m2 vs 30.83 ± 5.21 kg/m2), fasting C-peptide (0.81 ± 0.42 nmol/L vs 1.71 ± 0.80 nmol/L), insulin (76.81 ± 63.34 pmol/L vs 233.19 ± 164.51 pmol/L) and HOMA-IR index, despite similar fasting plasma glucose levels. Patients with pancreatogenic diabetes also had lower adrenomedullin levels (0.41 ± 0.25 ng/mL vs 0.63 ± 0.38 ng/mL) but higher adiponectin levels (13.08 ± 7.20 μg/mL vs 8.28 ± 4.01 μg/mL) and eGFR levels (100.53 ± 21.60 mL/min/1.73 m2 vs 85.14 ± 19.24 mL/min/1.73 m2). Finally, patients with pancreatogenic diabetes had significantly lower Type 3c diabetes index values. CONCLUSION Patients with pancreatogenic diabetes differ from patients with type 2 diabetes in anthropometric and laboratory parameters. The type 3c diabetes index had the highest discriminating value, above any single parameter.
Collapse
Affiliation(s)
- Anna Juza
- College of Medical Sciences, Institute of Medical Sciences, University of Rzeszów, Rzeszów 35-959, Poland
- Diabetic Outpatient Clinic & Department of Gastroenterology and Hepatology with the Subunit of Internal Diseases, University Clinical Hospital, Rzeszów 35-055, Poland
| | - Lilianna Kołodziej-Spirodek
- Diabetic Outpatient Clinic & Department of Gastroenterology and Hepatology with the Subunit of Internal Diseases, University Clinical Hospital, Rzeszów 35-055, Poland
| | - Krzysztof Gutkowski
- College of Medical Sciences, Institute of Medical Sciences, University of Rzeszów, Rzeszów 35-959, Poland
| | - Mariusz Partyka
- College of Medical Sciences, Institute of Medical Sciences, University of Rzeszów, Rzeszów 35-959, Poland
- Department of Internal Diseases, Nephrology and Endocrinology with the Nuclear Medicine Laboratory and the Dialysis Center & Endocrinology Outpatient Clinic, Clinical Provincial Hospital No. 2 in Rzeszów, Rzeszów 35-301, Poland
| | - Mariusz Dąbrowski
- College of Medical Sciences, Institute of Medical Sciences, University of Rzeszów, Rzeszów 35-959, Poland
| |
Collapse
|
2
|
Zilberg C, Ferguson AL, Lyons JG, Gupta R, Damian DL. The tumor immune microenvironment in primary cutaneous melanoma. Arch Dermatol Res 2025; 317:273. [PMID: 39825956 PMCID: PMC11742903 DOI: 10.1007/s00403-024-03758-8] [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: 07/19/2024] [Revised: 12/04/2024] [Accepted: 12/20/2024] [Indexed: 01/20/2025]
Abstract
Melanoma is an immunogenic tumor. The melanoma tumor immune microenvironment (TIME) is made up of a heterogenous mix of both immune and non-immune cells as well as a multitude of signaling molecules. The interactions between tumor cells, immune cells and signaling molecules affect tumor progression and therapeutic responses. Understanding the composition and function of the TIME in primary cutaneous melanoma is useful for prognostication and therapeutic decisions. This review provides an overview of the components of the TIME in primary cutaneous melanoma, and their influence on clinical outcomes.
Collapse
Affiliation(s)
- Catherine Zilberg
- The University of Sydney, NSW , Camperdown, 2050, Australia.
- Department of Tissue Pathology and Diagnostic Oncology, NSW Health Pathology, Royal Prince Alfred Hospital, Missenden Rd, Camperdown, NSW, 2050, Australia.
| | - Angela L Ferguson
- The University of Sydney, NSW , Camperdown, 2050, Australia
- Centenary Institute, The University of Sydney, Missenden Rd, Camperdown, NSW, 2050, Australia
| | - J Guy Lyons
- Centenary Institute, The University of Sydney, Missenden Rd, Camperdown, NSW, 2050, Australia
- Department of Dermatology, The University of Sydney at Royal Prince Alfred Hospital, Missenden Rd, NSW , Camperdown, 2050, Australia
| | - Ruta Gupta
- The University of Sydney, NSW , Camperdown, 2050, Australia
- Department of Tissue Pathology and Diagnostic Oncology, NSW Health Pathology, Royal Prince Alfred Hospital, Missenden Rd, Camperdown, NSW, 2050, Australia
| | - Diona L Damian
- Department of Dermatology, The University of Sydney at Royal Prince Alfred Hospital, Missenden Rd, NSW , Camperdown, 2050, Australia
- Melanoma Institute Australia, 40 Rocklands Rd, NSW, Wollstonecraft, 2065, Australia
| |
Collapse
|
3
|
Lee MJ, Cho JY, Bae S, Jung HS, Kang CM, Kim SH, Choi HJ, Lee CK, Kim H, Jo D, Paik YK. Inhibition of the Alternative Complement Pathway May Cause Secretion of Factor B, Enabling an Early Detection of Pancreatic Cancer. J Proteome Res 2024; 23:985-998. [PMID: 38306169 DOI: 10.1021/acs.jproteome.3c00695] [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] [Indexed: 02/03/2024]
Abstract
This study aims to elucidate the cellular mechanisms behind the secretion of complement factor B (CFB), known for its dual roles as an early biomarker for pancreatic ductal adenocarcinoma (PDAC) and as the initial substrate for the alternative complement pathway (ACP). Using parallel reaction monitoring analysis, we confirmed a consistent ∼2-fold increase in CFB expression in PDAC patients compared with that in both healthy donors (HD) and chronic pancreatitis (CP) patients. Elevated ACP activity was observed in CP and other benign conditions compared with that in HD and PDAC patients, suggesting a functional link between ACP and PDAC. Protein-protein interaction analyses involving key complement proteins and their regulatory factors were conducted using blood samples from PDAC patients and cultured cell lines. Our findings revealed a complex control system governing the ACP and its regulatory factors, including Kirsten rat sarcoma viral oncogene homolog (KRAS) mutation, adrenomedullin (AM), and complement factor H (CFH). Particularly, AM emerged as a crucial player in CFB secretion, activating CFH and promoting its predominant binding to C3b over CFB. Mechanistically, our data suggest that the KRAS mutation stimulates AM expression, enhancing CFH activity in the fluid phase through binding. This heightened AM-CFH interaction conferred greater affinity for C3b over CFB, potentially suppressing the ACP cascade. This sequence of events likely culminated in the preferential release of ductal CFB into plasma during the early stages of PDAC. (Data set ID PXD047043.).
Collapse
Affiliation(s)
- Min Jung Lee
- Yonsei Proteome Research Center, Yonsei University, Seoul 03722, South Korea
| | - Jin-Young Cho
- Yonsei Proteome Research Center, Yonsei University, Seoul 03722, South Korea
| | - Sumi Bae
- JW BioScience Corp., 38 Gwacheon-daero, Gwacheon-si, Gyeonggi-do 13840, South Korea
| | - Hye Soo Jung
- JW BioScience Corp., 38 Gwacheon-daero, Gwacheon-si, Gyeonggi-do 13840, South Korea
| | - Chang Moo Kang
- Department of Surgery, Division of HBP Surgery, Yonsei University College of Medicine, Seoul 03722, South Korea
| | - Sung Hyun Kim
- Department of Surgery, Division of HBP Surgery, Yonsei University College of Medicine, Seoul 03722, South Korea
| | - Hye Jin Choi
- Division of Medical Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul 03722, South Korea
| | - Choong-Kun Lee
- Division of Medical Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul 03722, South Korea
| | - Hoguen Kim
- Department of Pathology, Yonsei University College of Medicine, Seoul 03722, South Korea
| | - Daewoong Jo
- Cellivery R&D Institute, Cellivery Therapeutics, Inc., Seoul 03929, Korea
| | - Young-Ki Paik
- Yonsei Proteome Research Center, Yonsei University, Seoul 03722, South Korea
- Cellivery R&D Institute, Cellivery Therapeutics, Inc., Seoul 03929, Korea
| |
Collapse
|
4
|
Yang S, Huan R, Deng M, Luo T, Peng S, Xiong Y, Han G, Liu J, Zhang J, Tan Y. Pan-cancer analysis revealed prognosis value and immunological relevance of RAMPs. Heliyon 2024; 10:e24849. [PMID: 38317990 PMCID: PMC10838762 DOI: 10.1016/j.heliyon.2024.e24849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 12/09/2023] [Accepted: 01/16/2024] [Indexed: 02/07/2024] Open
Abstract
Whether receptor activity-modifying proteins (RAMPs) play a key role in human cancer prognosis and immunity remains unknown. We used data from the public databases, The Cancer Genome Atlas, Therapeutically Applicable Research to Generate Effective Treatments, and the Genotype-Tissue Expression project. We utilized bioinformatics methods, R software, and a variety of online databases to analyze RAMPs. In general, RAMPs were significantly and differentially expressed in multiple tumors, and RAMP expression was closely associated with prognosis, immune checkpoints, RNA-editing genes, tumor mutational burden, microsatellite instability, ploidy, and stemness indices. In addition, the expression of RAMPs is strongly correlated with tumor-infiltrating lymphocytes in human cancers. Moreover, the RAMP co-expression network is largely involved in many immune-related biological processes. Quantitative reverse transcription polymerase chain reaction and Western blot proved that RAMP3 was highly expressed in glioma, and RAMP3 promoted tumor proliferation and migration. RAMPs exhibit potential as prognostic and immune-related biomarkers in human cancers. Moreover, RAMPs can be potentially developed as therapeutic targets or used to enhance the efficacy of immunotherapy.
Collapse
Affiliation(s)
- Sha Yang
- Guizhou University Medical College, Guiyang, 550025, Guizhou Province, China
| | - Renzheng Huan
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Mei Deng
- Department of Neurosurgery, Guizhou Provincial People's Hospital, Guiyang, China
| | - Tao Luo
- Department of Neurosurgery, Guizhou Provincial People's Hospital, Guiyang, China
| | - Shuo Peng
- Department of Neurosurgery, Guizhou Provincial People's Hospital, Guiyang, China
| | - Yunbiao Xiong
- Department of Neurosurgery, Guizhou Provincial People's Hospital, Guiyang, China
| | - Guoqiang Han
- Department of Neurosurgery, Guizhou Provincial People's Hospital, Guiyang, China
| | - Jian Liu
- Guizhou University Medical College, Guiyang, 550025, Guizhou Province, China
- Department of Neurosurgery, Guizhou Provincial People's Hospital, Guiyang, China
| | - Jiqin Zhang
- Department of Anesthesiology, Guizhou Provincial People's Hospital, Guiyang, China
| | - Ying Tan
- Department of Neurosurgery, Guizhou Provincial People's Hospital, Guiyang, China
| |
Collapse
|
5
|
Expression profile of adrenomedullin and its specific receptors in liver tissues from patients with hepatocellular carcinoma and in tumorigenic cell line-secreted extracellular vesicles. Pathol Res Pract 2023; 243:154383. [PMID: 36827885 DOI: 10.1016/j.prp.2023.154383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 02/18/2023] [Indexed: 02/22/2023]
Abstract
The transcriptional profile of adrenomedullin (AM), a new metastasis-related factor involved in hepatocellular carcinoma (HCC), and its specific receptors (CLR, RAMP1, RAMP3) were evaluated in liver tissues of HCV-positive HCC subjects undergoing liver transplantation (LR) and in donors (LD). AM and its specific receptor expression were also assessed in extracellular vesicles (EVs) secreted by tumorigenic (HepG2) and non-tumorigenic (WRL68) cells by Real-Time PCR. AM expression resulted significantly elevated in LR concerning LD (p = 0.0038) and, for the first time, significantly higher levels in HCC patients as a function of clinical severity (MELD score), were observed. RAMP3 and CLR expression increased in LR as a function of clinical severity while RAMP1 decreased. Positive correlations were found among AM, its receptors, and apoptotic markers. No AM mRNA expression difference was observed between HepG2 and WRL68 EVs. RAMP1 and RAMP3 resulted lower in HepG2 concerning WRL68 while significantly higher levels were observed for CLR. While results at tissue level characterize AM as a regulator of carcinogenesis-tumor progression, those obtained in EVs do not indicate AM as a target candidate, neither as a pathological biomarker nor as a marker involved in cancer therapy.
Collapse
|
6
|
Expression of the Calcitonin Receptor-like Receptor (CALCRL) in Normal and Neoplastic Tissues. Int J Mol Sci 2023; 24:ijms24043960. [PMID: 36835377 PMCID: PMC9962437 DOI: 10.3390/ijms24043960] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 02/14/2023] [Accepted: 02/14/2023] [Indexed: 02/18/2023] Open
Abstract
Little information is available concerning protein expression of the calcitonin receptor-like receptor (CALCRL) at the protein level. Here, we developed a rabbit monoclonal antibody, 8H9L8, which is directed against human CALCRL but cross-reacts with the rat and mouse forms of the receptor. We confirmed antibody specificity via Western blot analyses and immunocytochemistry using the CALCRL-expressing neuroendocrine tumour cell line BON-1 and a CALCRL-specific small interfering RNA (siRNA). We then used the antibody for immunohistochemical analyses of various formalin-fixed, paraffin-embedded specimens of normal and neoplastic tissues. In nearly all tissue specimens examined, CALCRL expression was detected in the capillary endothelium, smooth muscles of the arterioles and arteries, and immune cells. Analyses of normal human, rat, and mouse tissues revealed that CALCRL was primarily present in distinct cell populations in the cerebral cortex; pituitary; dorsal root ganglia; epithelia, muscles, and glands of the larger bronchi; intestinal mucosa (particularly in enteroendocrine cells); intestinal ganglia; exocrine and endocrine pancreas; arteries, capillaries, and glomerular capillary loops in the kidneys; the adrenals; Leydig cells in the testicles; and syncytiotrophoblasts in the placenta. In the neoplastic tissues, CALCRL was predominantly expressed in thyroid carcinomas, parathyroid adenomas, small-cell lung cancers, large-cell neuroendocrine carcinomas of the lung, pancreatic neuroendocrine neoplasms, renal clear-cell carcinomas, pheochromocytomas, lymphomas, and melanomas. In these tumours with strong expression of CALCRL, the receptor may represent a useful target structure for future therapies.
Collapse
|
7
|
Taylor AJ, Panzhinskiy E, Orban PC, Lynn FC, Schaeffer DF, Johnson JD, Kopp JL, Verchere CB. Islet amyloid polypeptide does not suppress pancreatic cancer. Mol Metab 2023; 68:101667. [PMID: 36621763 PMCID: PMC9938314 DOI: 10.1016/j.molmet.2023.101667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 12/24/2022] [Accepted: 01/02/2023] [Indexed: 01/06/2023] Open
Abstract
OBJECTIVES Pancreatic cancer risk is elevated approximately two-fold in type 1 and type 2 diabetes. Islet amyloid polypeptide (IAPP) is an abundant beta-cell peptide hormone that declines with diabetes progression. IAPP has been reported to act as a tumour-suppressor in p53-deficient cancers capable of regressing tumour volumes. Given the decline of IAPP during diabetes development, we investigated the actions of IAPP in pancreatic ductal adenocarcinoma (PDAC; the most common form of pancreatic cancer) to determine if IAPP loss in diabetes may increase the risk of pancreatic cancer. METHODS PANC-1, MIA PaCa-2, and H1299 cells were treated with rodent IAPP, and the IAPP analogs pramlintide and davalintide, and assayed for changes in proliferation, death, and glycolysis. An IAPP-deficient mouse model of PDAC (Iapp-/-; Kras+/LSL-G12D; Trp53flox/flox; Ptf1a+/CreER) was generated for survival analysis. RESULTS IAPP did not impact glycolysis in MIA PaCa-2 cells, and did not impact cell death, proliferation, or glycolysis in PANC-1 cells or in H1299 cells, which were previously reported as IAPP-sensitive. Iapp deletion in Kras+/LSL-G12D; Trp53flox/flox; Ptf1a+/CreER mice had no effect on survival time to lethal tumour burden. CONCLUSIONS In contrast to previous reports, we find that IAPP does not function as a tumour suppressor. This suggests that loss of IAPP signalling likely does not increase the risk of pancreatic cancer in individuals with diabetes.
Collapse
Affiliation(s)
- Austin J Taylor
- BC Children's Hospital Research Institute, Vancouver, BC, Canada; Centre for Molecular Medicine and Therapeutics, Vancouver, BC, Canada; Department of Pathology and Laboratory Medicine, University of British Columbia, BC, Canada
| | - Evgeniy Panzhinskiy
- Life Sciences Institute, University of British Columbia, BC, Canada; Department of Biochemistry, University of British Columbia, BC, Canada
| | - Paul C Orban
- BC Children's Hospital Research Institute, Vancouver, BC, Canada; Centre for Molecular Medicine and Therapeutics, Vancouver, BC, Canada; Department of Surgery, University of British Columbia, BC, Canada
| | - Francis C Lynn
- BC Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Surgery, University of British Columbia, BC, Canada; Department of Cellular and Physiological Sciences, University of British Columbia, BC, Canada
| | - David F Schaeffer
- Department of Pathology and Laboratory Medicine, University of British Columbia, BC, Canada; Pancreas Centre BC, Vancouver, BC, Canada
| | - James D Johnson
- Life Sciences Institute, University of British Columbia, BC, Canada; Department of Cellular and Physiological Sciences, University of British Columbia, BC, Canada
| | - Janel L Kopp
- Life Sciences Institute, University of British Columbia, BC, Canada; Department of Cellular and Physiological Sciences, University of British Columbia, BC, Canada
| | - C Bruce Verchere
- BC Children's Hospital Research Institute, Vancouver, BC, Canada; Centre for Molecular Medicine and Therapeutics, Vancouver, BC, Canada; Department of Pathology and Laboratory Medicine, University of British Columbia, BC, Canada; Department of Surgery, University of British Columbia, BC, Canada.
| |
Collapse
|
8
|
Benyahia Z, Gaudy-Marqueste C, Berenguer-Daizé C, Chabane N, Dussault N, Cayol M, Vellutini C, Djemli A, Nanni I, Beaufils N, Mabrouk K, Grob JJ, Ouafik L. Adrenomedullin Secreted by Melanoma Cells Promotes Melanoma Tumor Growth through Angiogenesis and Lymphangiogenesis. Cancers (Basel) 2022; 14:cancers14235909. [PMID: 36497391 PMCID: PMC9738606 DOI: 10.3390/cancers14235909] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 12/03/2022] Open
Abstract
INTRODUCTION Metastatic melanoma is an aggressive tumor and can constitute a real therapeutic challenge despite the significant progress achieved with targeted therapies and immunotherapies, thus highlighting the need for the identification of new therapeutic targets. Adrenomedullin (AM) is a peptide with significant expression in multiple types of tumors and is multifunctional. AM impacts angiogenesis and tumor growth and binds to calcitonin receptor-like receptor/receptor activity-modifying protein 2 or 3 (CLR/RAMP2; CLR/RAMP3). METHODS In vitro and in vivo studies were performed to determine the functional role of AM in melanoma growth and tumor-associated angiogenesis and lymphangiogenesis. RESULTS In this study, AM and AM receptors were immunohistochemically localized in the tumoral compartment of melanoma tissue, suggesting that the AM system plays a role in melanoma growth. We used A375, SK-MEL-28, and MeWo cells, for which we demonstrate an expression of AM and its receptors; hypoxia induces the expression of AM in melanoma cells. The proliferation of A375 and SK-MEL-28 cells is decreased by anti-AM antibody (αAM) and anti-AMR antibodies (αAMR), supporting the fact that AM may function as a potent autocrine/paracrine growth factor for melanoma cells. Furthermore, migration and invasion of melanoma cells increased after treatment with AM and decreased after treatment with αAMR, thus indicating that melanoma cells are regulated by AM. Systemic administration of αAMR reduced neovascularization of in vivo Matrigel plugs containing melanoma cells, as demonstrated by reduced numbers of vessel structures, which suggests that AM is one of the melanoma cells-derived factors responsible for endothelial cell-like and pericyte recruitment in the construction of neovascularization. In vivo, αAMR therapy blocked angiogenesis and lymphangiogenesis and decreased proliferation in MeWo xenografts, thereby resulting in tumor regression. Histological examination of αAMR-treated tumors showed evidence of the disruption of tumor vascularity, with depletion of vascular endothelial cells and a significant decrease in lymphatic endothelial cells. CONCLUSIONS The expression of AM by melanoma cells promotes tumor growth and neovascularization by supplying/amplifying signals for neoangiogenesis and lymphangiogenesis.
Collapse
Affiliation(s)
- Zohra Benyahia
- Aix Marseille Univ, CNRS, INP, Inst Neurophysiopathol, 13005 Marseille, France
| | - Caroline Gaudy-Marqueste
- Aix Marseille Univ, APHM, CHU Timone, Service de Dermatologie et de Cancérologie Cutanée, 13005 Marseille, France
| | | | - Norhimane Chabane
- Aix Marseille Univ, CNRS, INP, Inst Neurophysiopathol, 13005 Marseille, France
| | - Nadège Dussault
- Aix Marseille Univ, CNRS, INP, Inst Neurophysiopathol, 13005 Marseille, France
| | - Mylène Cayol
- Aix Marseille Univ, CNRS, INP, Inst Neurophysiopathol, 13005 Marseille, France
| | - Christine Vellutini
- Aix Marseille Univ, CNRS, INP, Inst Neurophysiopathol, 13005 Marseille, France
| | - Amina Djemli
- Aix Marseille Univ, APHM, CHU Nord, Service D’anatomopathologie, 13015 Marseille, France
| | - Isabelle Nanni
- Aix Marseille Univ, APHM, CHU Nord, Service D’Onco-Biologie, 13015 Marseille, France
| | - Nathalie Beaufils
- Aix Marseille Univ, APHM, CHU Nord, Service D’Onco-Biologie, 13015 Marseille, France
| | - Kamel Mabrouk
- Aix Marseille Univ, CNRS, ICR, Institut de Chimie Radicalaire, 13013 Marseille, France
| | - Jean-Jacques Grob
- Aix Marseille Univ, APHM, CHU Timone, Service de Dermatologie et de Cancérologie Cutanée, 13005 Marseille, France
| | - L’Houcine Ouafik
- Aix Marseille Univ, CNRS, INP, Inst Neurophysiopathol, 13005 Marseille, France
- Aix Marseille Univ, APHM, CHU Nord, Service D’Onco-Biologie, 13015 Marseille, France
- Correspondence: ; Tel.: +33-491324447
| |
Collapse
|
9
|
Wang L, Liu Z, Liang R, Wang W, Zhu R, Li J, Xing Z, Weng S, Han X, Sun YL. Comprehensive machine-learning survival framework develops a consensus model in large-scale multicenter cohorts for pancreatic cancer. eLife 2022; 11:e80150. [PMID: 36282174 PMCID: PMC9596158 DOI: 10.7554/elife.80150] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 10/15/2022] [Indexed: 11/13/2022] Open
Abstract
As the most aggressive tumor, the outcome of pancreatic cancer (PACA) has not improved observably over the last decade. Anatomy-based TNM staging does not exactly identify treatment-sensitive patients, and an ideal biomarker is urgently needed for precision medicine. Based on expression files of 1280 patients from 10 multicenter cohorts, we screened 32 consensus prognostic genes. Ten machine-learning algorithms were transformed into 76 combinations, of which we selected the optimal algorithm to construct an artificial intelligence-derived prognostic signature (AIDPS) according to the average C-index in the nine testing cohorts. The results of the training cohort, nine testing cohorts, Meta-Cohort, and three external validation cohorts (290 patients) consistently indicated that AIDPS could accurately predict the prognosis of PACA. After incorporating several vital clinicopathological features and 86 published signatures, AIDPS exhibited robust and dramatically superior predictive capability. Moreover, in other prevalent digestive system tumors, the nine-gene AIDPS could still accurately stratify the prognosis. Of note, our AIDPS had important clinical implications for PACA, and patients with low AIDPS owned a dismal prognosis, higher genomic alterations, and denser immune cell infiltrates as well as were more sensitive to immunotherapy. Meanwhile, the high AIDPS group possessed observably prolonged survival, and panobinostat may be a potential agent for patients with high AIDPS. Overall, our study provides an attractive tool to further guide the clinical management and individualized treatment of PACA.
Collapse
Affiliation(s)
- Libo Wang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Institute of Hepatobiliary and Pancreatic Diseases, Zhengzhou UniversityZhengzhouChina
- Zhengzhou Basic and Clinical Key Laboratory of Hepatopancreatobiliary DiseasesZhengzhouChina
| | - Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Ruopeng Liang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Institute of Hepatobiliary and Pancreatic Diseases, Zhengzhou UniversityZhengzhouChina
- Zhengzhou Basic and Clinical Key Laboratory of Hepatopancreatobiliary DiseasesZhengzhouChina
| | - Weijie Wang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Institute of Hepatobiliary and Pancreatic Diseases, Zhengzhou UniversityZhengzhouChina
- Zhengzhou Basic and Clinical Key Laboratory of Hepatopancreatobiliary DiseasesZhengzhouChina
| | - Rongtao Zhu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Institute of Hepatobiliary and Pancreatic Diseases, Zhengzhou UniversityZhengzhouChina
- Zhengzhou Basic and Clinical Key Laboratory of Hepatopancreatobiliary DiseasesZhengzhouChina
| | - Jian Li
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Institute of Hepatobiliary and Pancreatic Diseases, Zhengzhou UniversityZhengzhouChina
- Zhengzhou Basic and Clinical Key Laboratory of Hepatopancreatobiliary DiseasesZhengzhouChina
| | - Zhe Xing
- Department of Neurosurgery, The Fifth Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Siyuan Weng
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Yu-ling Sun
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Institute of Hepatobiliary and Pancreatic Diseases, Zhengzhou UniversityZhengzhouChina
- Zhengzhou Basic and Clinical Key Laboratory of Hepatopancreatobiliary DiseasesZhengzhouChina
| |
Collapse
|
10
|
Song C, Pan S, Li D, Hao B, Lu Z, Lai K, Li N, Geng Q. Comprehensive analysis reveals the potential value of inflammatory response genes in the prognosis, immunity, and drug sensitivity of lung adenocarcinoma. BMC Med Genomics 2022; 15:198. [PMID: 36117156 PMCID: PMC9484176 DOI: 10.1186/s12920-022-01340-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 08/16/2022] [Indexed: 11/18/2022] Open
Abstract
Background Although the relationship between inflammatory response and tumor has been gradually recognized, the potential implications of of inflammatory response genes in lung adenocarcinoma (LUAD) remains poorly investigated. Methods RNA sequencing and clinical data were obtained from multiple independent datasets (GSE29013, GSE30219, GSE31210, GSE37745, GSE42127, GSE50081, GSE68465, GSE72094, TCGA and GTEx). Unsupervised clustering analysis was used to identify different tumor subtypes, and LASSO and Cox regression analysis were applied to construct a novel scoring tool. We employed multiple algorithms (ssGSEA, CIBERSORT, MCP counter, and ESTIMATE) to better characterize the LUAD tumor microenvironment (TME) and immune landscapes. GSVA and Metascape analysis were performed to investigate the biological processes and pathway activity. Furthermore, ‘pRRophetic’ R package was used to evaluate the half inhibitory concentration (IC50) of each sample to infer drug sensitivity. Results We identified three distinct tumor subtypes, which were related to different clinical outcomes, biological pathways, and immune characteristics. A scoring tool called inflammatory response gene score (IRGS) was established and well validated in multiple independent cohorts, which could well divide patients into two subgroups with significantly different prognosis. High IRGS patients, characterized by increased genomic variants and mutation burden, presented a worse prognosis, and might show a more favorable response to immunotherapy and chemotherapy. Additionally, based on the cross-talk between TNM stage, IRGS and patients clinical outcomes, we redefined the LUAD stage, which was called ‘IRGS-Stage’. The novel staging system could distinguish patients with different prognosis, with better predictive ability than the conventional TNM staging. Conclusions Inflammatory response genes present important potential value in the prognosis, immunity and drug sensitivity of LUAD. The proposed IRGS and IRGS-Stage may be promising biomarkers for estimating clinical outcomes in LUAD patients. Supplementary Information The online version contains supplementary material available at 10.1186/s12920-022-01340-7.
Collapse
|
11
|
Receptor Activity Modifying Protein RAMP Sub-Isoforms and Their Functional Differentiation, Which Regulates Functional Diversity of Adrenomedullin. BIOLOGY 2022; 11:biology11050788. [PMID: 35625516 PMCID: PMC9138304 DOI: 10.3390/biology11050788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/19/2022] [Accepted: 05/20/2022] [Indexed: 11/17/2022]
Abstract
AM knockout (AM-/-) and RAMP2 knockout (RAMP2-/-) proved lethal for mice due to impaired embryonic vascular development. Although most vascular endothelial cell-specific RAMP2 knockout (E-RAMP2-/-) mice also died during the perinatal period, a few E-RAMP2-/- mice reached adulthood. Adult E-RAMP2-/- mice developed spontaneous organ damage associated with vascular injury. In contrast, adult RAMP3 knockout (RAMP3-/-) mice showed exacerbated postoperative lymphedema with abnormal lymphatic drainage. Thus, RAMP2 is essential for vascular development and homeostasis and RAMP3 is essential for lymphatic vessel function. Cardiac myocyte-specific RAMP2 knockout mice showed early onset of heart failure as well as abnormal mitochondrial morphology and function, whereas RAMP3-/- mice exhibited abnormal cardiac lymphatics and a delayed onset of heart failure. Thus, RAMP2 is essential for maintaining cardiac mitochondrial function, while RAMP3 is essential for cardiac lymphangiogenesis. Transplantation of cancer cells into drug-inducible vascular endothelial cell-specific RAMP2 knockout mice resulted in enhanced metastasis to distant organs, whereas metastasis was suppressed in RAMP3-/- mice. RAMP2 suppresses cancer metastasis by maintaining vascular homeostasis and inhibiting vascular inflammation and pre-metastatic niche formation, while RAMP3 promotes cancer metastasis via malignant transformation of cancer-associated fibroblasts. Focusing on the diverse physiological functions of AM and the functional differentiation of RAMP2 and RAMP3 may lead to the development of novel therapeutic strategies.
Collapse
|
12
|
Identification of an Inflammatory Response-Related Gene Signature to Predict Survival and Immune Status in Glioma Patients. J Immunol Res 2022; 2022:8972730. [PMID: 35647198 PMCID: PMC9132661 DOI: 10.1155/2022/8972730] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 03/14/2022] [Accepted: 04/21/2022] [Indexed: 12/26/2022] Open
Abstract
Background Glioma is the most common primary brain tumor with high mortality and poor outcomes. As a hallmark of cancers, inflammatory responses are crucial for their progression. The present study is aimed at exploring the prognostic value of inflammatory response-related genes (IRRGs) and constructing a prognostic IRRG signature for gliomas. Materials and Methods We investigated the relationship between IRRGs and gliomas by integrating the transcriptomic data for gliomas from public databases. Differentially expressed IRRGs (DE-IRRGs) were identified in the GSE4290 cohort. Further, univariate, least absolute shrinkage and selection operator, and multivariate Cox regression analyses were conducted to construct an IRRG signature using The Cancer Genome Atlas (TCGA) cohort. Gliomas from the Chinese Glioma Genome Atlas (CGGA) cohort were employed for independent validation. The performance of gene signature was assessed by survival and receiver operating characteristic curve analyses. The differences in clinical correlations, immune infiltrate types, immunotherapeutic response predictions, and pathway enrichment among subgroups were investigated via bioinformatic algorithms. Results In total, 37 DE-IRRGs were determined, of which 31 were found to be associated with survival. Ultimately, eight genes were retained to construct an IRRG signature that further classified glioma patients into two groups; the high-risk group suffered a poorer outcome as compared to the low-risk group. Furthermore, the high-risk group was significantly correlated with several risk factors, including older age, higher tumor grade, IDH wild type, 1p19q noncodel, and MGMT unmethylation. The nomogram was constructed by integrating the risk scores and other independent clinical characteristics. Moreover, the high-risk group had a greater immune infiltration and was most likely to benefit from immunotherapy. Gene set enrichment analysis suggested that immune and oncogenic pathways were enriched in high-risk glioma patients. Conclusion We constructed a signature composed of eight IRRGs for gliomas, which could effectively predict survival and guide decision-making for treatment.
Collapse
|
13
|
Sigaud R, Dussault N, Berenguer-Daizé C, Vellutini C, Benyahia Z, Cayol M, Parat F, Mabrouk K, Vázquez R, Riveiro ME, Metellus P, Ouafik L. Role of the Tyrosine Phosphatase SHP-2 in Mediating Adrenomedullin Proangiogenic Activity in Solid Tumors. Front Oncol 2021; 11:753244. [PMID: 34692535 PMCID: PMC8531523 DOI: 10.3389/fonc.2021.753244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 09/14/2021] [Indexed: 11/17/2022] Open
Abstract
VE-cadherin is an essential adhesion molecule in endothelial adherens junctions, and the integrity of these complexes is thought to be regulated by VE-cadherin tyrosine phosphorylation. We have previously shown that adrenomedullin (AM) blockade correlates with elevated levels of phosphorylated VE-cadherin (pVE-cadherinY731) in endothelial cells, associated with impaired barrier function and a persistent increase in vascular endothelial cell permeability. However, the mechanism underlying this effect is unknown. In this article, we demonstrate that the AM-mediated dephosphorylation of pVE-cadherinY731 takes place through activation of the tyrosine phosphatase SHP-2, as judged by the rise of its active fraction phosphorylated at tyrosine 542 (pSHP-2Y542) in HUVECs and glioblastoma-derived-endothelial cells. Both pre-incubation of HUVECs with SHP-2 inhibitors NSC-87877 and SHP099 and SHP-2 silencing hindered AM-induced dephosphorylation of pVE-cadherinY731 in a dose dependent-manner, showing the role of SHP-2 in the regulation of endothelial cell contacts. Furthermore, SHP-2 inhibition impaired AM-induced HUVECs differentiation into cord-like structures in vitro and impeded AM-induced neovascularization in in vivo Matrigel plugs bioassays. Subcutaneously transplanted U87-glioma tumor xenograft mice treated with AM-receptors-blocking antibodies showed a decrease in pSHP-2Y542 associated with VE-cadherin in nascent tumor vasculature when compared to control IgG-treated xenografts. Our findings show that AM acts on VE-cadherin dynamics through pSHP-2Y542 to finally modulate cell-cell junctions in the angiogenesis process, thereby promoting a stable and functional tumor vasculature.
Collapse
Affiliation(s)
- Romain Sigaud
- Aix Marseille University, Centre National de la Recherche Scientifique (CNRS), Institut de Neurophysiopathologie( INP), Inst Neurophysiopathol, Marseille, France
| | - Nadège Dussault
- Aix Marseille University, Centre National de la Recherche Scientifique (CNRS), Institut de Neurophysiopathologie( INP), Inst Neurophysiopathol, Marseille, France
| | - Caroline Berenguer-Daizé
- Aix Marseille University, Centre National de la Recherche Scientifique (CNRS), Institut de Neurophysiopathologie( INP), Inst Neurophysiopathol, Marseille, France
| | - Christine Vellutini
- Aix Marseille University, Centre National de la Recherche Scientifique (CNRS), Institut de Neurophysiopathologie( INP), Inst Neurophysiopathol, Marseille, France
| | - Zohra Benyahia
- Aix Marseille University, Centre National de la Recherche Scientifique (CNRS), Institut de Neurophysiopathologie( INP), Inst Neurophysiopathol, Marseille, France
| | - Mylène Cayol
- Aix Marseille University, Centre National de la Recherche Scientifique (CNRS), Institut de Neurophysiopathologie( INP), Inst Neurophysiopathol, Marseille, France
| | - Fabrice Parat
- Aix Marseille University, Centre National de la Recherche Scientifique (CNRS), Institut de Neurophysiopathologie( INP), Inst Neurophysiopathol, Marseille, France
| | - Kamel Mabrouk
- Aix Marseille University, CNRS, Institut de Chimie Radicalaire (ICR), Unité Mixte de Recherche (UMR) 7273 Chimie Radicalaire Organique et Polymères de Spécialité (CROPS), Marseille, France
| | - Ramiro Vázquez
- Preclinical Department, Early Drug Development Group (E2DG), Boulogne-Billancourt, France.,Center for Genomic Science of Istituto Italiano di Tecnologia, Center for Genomic Science, European School of Molecular Medicine (IIT@SEMM), Fondazione Istituto Italiano di Tecnologia (IIT), Milan, Italy
| | - Maria E Riveiro
- Preclinical Department, Early Drug Development Group (E2DG), Boulogne-Billancourt, France
| | - Philippe Metellus
- Aix Marseille University, Centre National de la Recherche Scientifique (CNRS), Institut de Neurophysiopathologie( INP), Inst Neurophysiopathol, Marseille, France.,Centre Hospitalier Clairval, Département de Neurochirurgie, Marseille, France
| | - L'Houcine Ouafik
- Aix Marseille University, Centre National de la Recherche Scientifique (CNRS), Institut de Neurophysiopathologie( INP), Inst Neurophysiopathol, Marseille, France.,Assistance Publique Hôpitaux de Marseille (APHM), Centre Hospitalo Universitaire (CHU) Nord, Service d'OncoBiologie, Marseille, France
| |
Collapse
|
14
|
Neufeld L, Yeini E, Reisman N, Shtilerman Y, Ben-Shushan D, Pozzi S, Madi A, Tiram G, Eldar-Boock A, Ferber S, Grossman R, Ram Z, Satchi-Fainaro R. Microengineered perfusable 3D-bioprinted glioblastoma model for in vivo mimicry of tumor microenvironment. SCIENCE ADVANCES 2021; 7:eabi9119. [PMID: 34407932 PMCID: PMC8373143 DOI: 10.1126/sciadv.abi9119] [Citation(s) in RCA: 105] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 06/28/2021] [Indexed: 05/04/2023]
Abstract
Many drugs show promising results in laboratory research but eventually fail clinical trials. We hypothesize that one main reason for this translational gap is that current cancer models are inadequate. Most models lack the tumor-stroma interactions, which are essential for proper representation of cancer complexed biology. Therefore, we recapitulated the tumor heterogenic microenvironment by creating fibrin glioblastoma bioink consisting of patient-derived glioblastoma cells, astrocytes, and microglia. In addition, perfusable blood vessels were created using a sacrificial bioink coated with brain pericytes and endothelial cells. We observed similar growth curves, drug response, and genetic signature of glioblastoma cells grown in our 3D-bioink platform and in orthotopic cancer mouse models as opposed to 2D culture on rigid plastic plates. Our 3D-bioprinted model could be the basis for potentially replacing cell cultures and animal models as a powerful platform for rapid, reproducible, and robust target discovery; personalized therapy screening; and drug development.
Collapse
Affiliation(s)
- Lena Neufeld
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Eilam Yeini
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Noa Reisman
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Yael Shtilerman
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Dikla Ben-Shushan
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Sabina Pozzi
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Asaf Madi
- Department of Pathology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Galia Tiram
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Anat Eldar-Boock
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Shiran Ferber
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Rachel Grossman
- Department of Neurosurgery, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Zvi Ram
- Department of Neurosurgery, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Ronit Satchi-Fainaro
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel.
- Sagol School of Neurosciences, Tel Aviv University, Tel Aviv 69978, Israel
| |
Collapse
|
15
|
Patel JC, Singh A, Tulswani R, Sharma YK, Khurana P, Ragumani S. Identification of VEGFA-centric temporal hypoxia-responsive dynamic cardiopulmonary network biomarkers. Life Sci 2021; 281:119718. [PMID: 34147483 DOI: 10.1016/j.lfs.2021.119718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 05/31/2021] [Accepted: 06/08/2021] [Indexed: 10/21/2022]
Abstract
AIMS Hypoxia, a pathophysiological condition, is profound in several cardiopulmonary diseases (CPD). Every individual's lethality to a hypoxia state differs in terms of hypoxia exposure time, dosage units and dependent on the individual's genetic makeup. Most of the proposed markers for CPD were generally aim to distinguish disease samples from normal samples. Although, as per the 2018 GOLD guidelines, clinically useful biomarkers for several cardio pulmonary disease patients in stable condition have yet to be identified. We attempt to address these key issues through the identification of Dynamic Network Biomarkers (DNB) to detect hypoxia induced early warning signals of CPD before the catastrophic deterioration. MATERIALS AND METHODS The human microvascular endothelial tissues microarray datasets (GSE11341) of lung and cardiac expose to hypoxia (1% O2) for 3, 24 and 48 h were retrieved from the public repository. The time dependent differentially expressed genes were subjected to tissue specificity and promoter analysis to filtrate the noise levels in the networks and to dissect the tissue specific hypoxia induced genes. These filtered out genes were used to construct the dynamic segmentation networks. The hypoxia induced dynamic differentially expressed genes were validated in the lung and heart tissues of male rats. These rats were exposed to hypobaric hypoxia (simulated altitude of 25,000 or PO2 - 282 mm of Hg) progressively for 3, 24 and 48 h. KEY FINDINGS To identify the temporal key genes regulated in hypoxia, we ranked the dominant genes based on their consolidated topological features from tissue specific networks, time dependent networks and dynamic networks. Overall topological ranking described VEGFA as a single node dynamic hub and strongly communicated with tissue specific genes to carry forward their tissue specific information. We named this type of VEGFAcentric dynamic networks as "V-DNBs". As a proof of principle, our methodology helped us to identify the V-DNBs specific for lung and cardiac tissues namely V-DNBL and V-DNBC respectively. SIGNIFICANCE Our experimental studies identified VEGFA, SLC2A3, ADM and ENO2 as the minimum and sufficient candidates of V-DNBL. The dynamic expression patterns could be readily exploited to capture the pre disease state of hypoxia induced pulmonary vascular remodelling. Whereas in V-DNBC the minimum and sufficient candidates are VEGFA, SCL2A3, ADM, NDRG1, ENO2 and BHLHE40. The time dependent single node expansion indicates V-DNBC could also be the pre disease state pathological hallmark for hypoxia-associated cardiovascular remodelling. The network cross-talk and expression pattern between V-DNBL and V-DNBC are completely distinct. On the other hand, the great clinical advantage of V-DNBs for pre disease predictions, a set of samples during the healthy condition should suffice. Future clinical studies might further shed light on the predictive power of V-DNBs as prognostic and diagnostic biomarkers for CPD.
Collapse
Affiliation(s)
- Jai Chand Patel
- Defence Institute of Physiology and Allied Sciences, Defence Research and Development Organization, Lucknow Road, Timarpur, Delhi, India
| | - Ajeet Singh
- Defence Institute of Physiology and Allied Sciences, Defence Research and Development Organization, Lucknow Road, Timarpur, Delhi, India
| | - Rajkumar Tulswani
- Defence Institute of Physiology and Allied Sciences, Defence Research and Development Organization, Lucknow Road, Timarpur, Delhi, India
| | - Yogendra Kumar Sharma
- Defence Institute of Physiology and Allied Sciences, Defence Research and Development Organization, Lucknow Road, Timarpur, Delhi, India
| | - Pankaj Khurana
- Defence Institute of Physiology and Allied Sciences, Defence Research and Development Organization, Lucknow Road, Timarpur, Delhi, India
| | - Sugadev Ragumani
- Defence Institute of Physiology and Allied Sciences, Defence Research and Development Organization, Lucknow Road, Timarpur, Delhi, India.
| |
Collapse
|
16
|
Adrenomedullin Is a Diagnostic and Prognostic Biomarker for Acute Intracerebral Hemorrhage. Curr Issues Mol Biol 2021; 43:324-334. [PMID: 34208106 PMCID: PMC8928941 DOI: 10.3390/cimb43010027] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/07/2021] [Accepted: 06/09/2021] [Indexed: 12/17/2022] Open
Abstract
Hemorrhagic stroke remains an important health challenge. Adrenomedullin (AM) is a vasoactive peptide with an important role in cardiovascular diseases, including stroke. Serum AM and nitrate-nitrite and S-nitroso compounds (NOx) levels were measured and compared between healthy volunteers (n = 50) and acute hemorrhagic stroke patients (n = 64). Blood samples were taken at admission (d0), 24 h later (d1), and after 7 days or at the time of hospital discharge (d7). Neurological severity (NIHSS) and functional prognosis (mRankin) were measured as clinical outcomes. AM levels were higher in stroke patients at all times when compared with healthy controls (p < 0.0001). A receiving operating characteristic curve analysis identified that AM levels at admission > 69.0 pg/mL had a great value as a diagnostic biomarker (area under the curve = 0.89, sensitivity = 80.0%, specificity = 100%). Furthermore, patients with a favorable outcome (NIHSS ≤ 3; mRankin ≤ 2) experienced an increase in AM levels from d0 to d1, and a decrease from d1 to d7, whereas patients with unfavorable outcome had no significant changes over time. NOx levels were lower in patients at d0 (p = 0.04) and d1 (p < 0.001) than in healthy controls. In conclusion, AM levels may constitute a new diagnostic and prognostic biomarker for this disease, and identify AM as a positive mediator for hemorrhagic stroke resolution.
Collapse
|
17
|
Iriarte A, Ochoa-Callejero L, García-Sanmartín J, Cerdà P, Garrido P, Narro-Íñiguez J, Mora-Luján JM, Jucglà A, Sánchez-Corral MA, Cruellas F, Gamundi E, Ribas J, Castellote J, Viñals F, Martínez A, Riera-Mestre A. Adrenomedullin as a potential biomarker involved in patients with hereditary hemorrhagic telangiectasia. Eur J Intern Med 2021; 88:89-95. [PMID: 33888392 DOI: 10.1016/j.ejim.2021.03.039] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/28/2021] [Accepted: 03/30/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND Adrenomedullin (AM) is a vasoactive peptide mostly secreted by endothelial cells with an important role in preserving endothelial integrity. The relationship between AM and hereditary hemorrhagic telangiectasia (HHT) is unknown. We aimed to compare the serum levels and tissue expression of AM between HHT patients and controls. METHODS Serum AM levels were measured by radioimmunoassay and compared between control and HHT groups. AM levels were also compared among HHT subgroups according to clinical characteristics. The single nucleotide polymorphism (SNP) rs4910118 was assessed by restriction analysis and sequencing. AM immunohistochemistry was performed on biopsies of cutaneous telangiectasia from eight HHT patients and on the healthy skin from five patients in the control group. RESULTS Forty-five HHT patients and 50 healthy controls were included, mean age (SD) was 50.7 (14.9) years and 46.4 (9.9) years (p = 0.102), respectively. HHT patients were mostly female (60% vs 38%, p = 0.032). Median [Q1-Q3] serum AM levels were 68.3 [58.1-80.6] pg/mL in the HHT group and 47.7 [43.2-53.8] pg/mL in controls (p<0.001), with an optimal AM cut-off according to Youden's J statistic of 55.32 pg/mL (J:0.729). Serum AM levels were similar in the HHT subgroups. No patient with HHT had the SNP rs4910118. AM immunoreactivity was found with high intensity in the abnormal blood vessels of HHT biopsies. CONCLUSIONS We detected higher AM serum levels and tissue expression in patients with HHT than in healthy controls. The role of AM in HHT, and whether AM may constitute a novel biomarker and therapeutic target, needs further investigation.
Collapse
Affiliation(s)
- A Iriarte
- HHT Unit. Hospital Universitari de Bellvitge, Barcelona Spain; Internal Medicine Department. Hospital Universitari de Bellvitge, Barcelona Spain; Bellvitge Biomedical Research Institute (IDIBELL), Barcelona Spain
| | - L Ochoa-Callejero
- Angiogenesis Group, Oncology Area, Center for Biomedical Research of La Rioja (CIBIR), Logroño Spain
| | - J García-Sanmartín
- Angiogenesis Group, Oncology Area, Center for Biomedical Research of La Rioja (CIBIR), Logroño Spain
| | - P Cerdà
- HHT Unit. Hospital Universitari de Bellvitge, Barcelona Spain; Internal Medicine Department. Hospital Universitari de Bellvitge, Barcelona Spain; Bellvitge Biomedical Research Institute (IDIBELL), Barcelona Spain
| | - P Garrido
- Angiogenesis Group, Oncology Area, Center for Biomedical Research of La Rioja (CIBIR), Logroño Spain
| | - J Narro-Íñiguez
- Angiogenesis Group, Oncology Area, Center for Biomedical Research of La Rioja (CIBIR), Logroño Spain
| | - J M Mora-Luján
- HHT Unit. Hospital Universitari de Bellvitge, Barcelona Spain; Internal Medicine Department. Hospital Universitari de Bellvitge, Barcelona Spain; Bellvitge Biomedical Research Institute (IDIBELL), Barcelona Spain
| | - A Jucglà
- HHT Unit. Hospital Universitari de Bellvitge, Barcelona Spain; Bellvitge Biomedical Research Institute (IDIBELL), Barcelona Spain; Dermatology Department. Hospital Universitari de Bellvitge, Barcelona Spain
| | - M A Sánchez-Corral
- HHT Unit. Hospital Universitari de Bellvitge, Barcelona Spain; Bellvitge Biomedical Research Institute (IDIBELL), Barcelona Spain; Cardiology Department. Hospital Universitari de Bellvitge, Barcelona Spain
| | - F Cruellas
- HHT Unit. Hospital Universitari de Bellvitge, Barcelona Spain; Bellvitge Biomedical Research Institute (IDIBELL), Barcelona Spain; Otorhinolaryngology Department. Hospital Universitari de Bellvitge, Barcelona Spain
| | - E Gamundi
- Hematology Department. Hospital Universitari de Bellvitge, Barcelona Spain
| | - J Ribas
- HHT Unit. Hospital Universitari de Bellvitge, Barcelona Spain; Bellvitge Biomedical Research Institute (IDIBELL), Barcelona Spain; Pneumology Department. Hospital Universitari de Bellvitge, Barcelona Spain
| | - J Castellote
- HHT Unit. Hospital Universitari de Bellvitge, Barcelona Spain; Bellvitge Biomedical Research Institute (IDIBELL), Barcelona Spain; Liver Transplant Unit, Gastroenterology Department. Hospital Universitari de Bellvitge, Barcelona Spain; Physiological Sciences Department. Faculty of Medicine and Health Sciences. Universitat de Barcelona, Barcelona, Spain
| | - F Viñals
- Physiological Sciences Department. Faculty of Medicine and Health Sciences. Universitat de Barcelona, Barcelona, Spain; Program Against Cancer Therapeutic Resistance, Institut Catala d'Oncologia, Hospital Duran i Reynals, Barcelona Spain; Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
| | - A Martínez
- Angiogenesis Group, Oncology Area, Center for Biomedical Research of La Rioja (CIBIR), Logroño Spain
| | - A Riera-Mestre
- HHT Unit. Hospital Universitari de Bellvitge, Barcelona Spain; Internal Medicine Department. Hospital Universitari de Bellvitge, Barcelona Spain; Bellvitge Biomedical Research Institute (IDIBELL), Barcelona Spain; Faculty of Medicine and Health Sciences. Universitat de Barcelona, Barcelona, Spain.
| |
Collapse
|
18
|
Vázquez R, Riveiro ME, Berenguer-Daizé C, O'Kane A, Gormley J, Touzelet O, Rezai K, Bekradda M, Ouafik L. Targeting Adrenomedullin in Oncology: A Feasible Strategy With Potential as Much More Than an Alternative Anti-Angiogenic Therapy. Front Oncol 2021; 10:589218. [PMID: 33489885 PMCID: PMC7815935 DOI: 10.3389/fonc.2020.589218] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 11/02/2020] [Indexed: 12/18/2022] Open
Abstract
The development, maintenance and metastasis of solid tumors are highly dependent on the formation of blood and lymphatic vessels from pre-existing ones through a series of processes that are respectively known as angiogenesis and lymphangiogenesis. Both are mediated by specific growth-stimulating molecules, such as the vascular endothelial growth factor (VEGF) and adrenomedullin (AM), secreted by diverse cell types which involve not only the cancerogenic ones, but also those constituting the tumor stroma (i.e., macrophages, pericytes, fibroblasts, and endothelial cells). In this sense, anti-angiogenic therapy represents a clinically-validated strategy in oncology. Current therapeutic approaches are mainly based on VEGF-targeting agents, which, unfortunately, are usually limited by toxicity and/or tumor-acquired resistance. AM is a ubiquitous peptide hormone mainly secreted in the endothelium with an important involvement in blood vessel development and cardiovascular homeostasis. In this review, we will introduce the state-of-the-art in terms of AM physiology, while putting a special focus on its pro-tumorigenic role, and discuss its potential as a therapeutic target in oncology. A large amount of research has evidenced AM overexpression in a vast majority of solid tumors and a correlation between AM levels and disease stage, progression and/or vascular density has been observed. The analysis presented here indicates that the involvement of AM in the pathogenesis of cancer arises from: 1) direct promotion of cell proliferation and survival; 2) increased vascularization and the subsequent supply of nutrients and oxygen to the tumor; 3) and/or alteration of the cell phenotype into a more aggressive one. Furthermore, we have performed a deep scrutiny of the pathophysiological prominence of each of the AM receptors (AM1 and AM2) in different cancers, highlighting their differential locations and functions, as well as regulatory mechanisms. From the therapeutic point of view, we summarize here an exhaustive series of preclinical studies showing a reduction of tumor angiogenesis, metastasis and growth following treatment with AM-neutralizing antibodies, AM receptor antagonists, or AM receptor interference. Anti-AM therapy is a promising strategy to be explored in oncology, not only as an anti-angiogenic alternative in the context of acquired resistance to VEGF treatment, but also as a potential anti-metastatic approach.
Collapse
Affiliation(s)
- Ramiro Vázquez
- Preclinical Department, Early Drug Development Group (E2DG), Boulogne-Billancourt, France.,Center for Genomic Science of IIT@SEMM, Fondazione Istituto Italiano di Tecnologia (IIT), Milan, Italy
| | - Maria E Riveiro
- Preclinical Department, Early Drug Development Group (E2DG), Boulogne-Billancourt, France
| | | | - Anthony O'Kane
- Discovery and Scientific Affairs Department, Fusion Antibodies plc., Belfast, United Kingdom
| | - Julie Gormley
- Discovery and Scientific Affairs Department, Fusion Antibodies plc., Belfast, United Kingdom
| | - Olivier Touzelet
- Discovery and Scientific Affairs Department, Fusion Antibodies plc., Belfast, United Kingdom
| | - Keyvan Rezai
- Department of Radio-Pharmacology, Institute Curie-René Huguenin Hospital, Saint-Cloud, France
| | - Mohamed Bekradda
- Preclinical Department, Early Drug Development Group (E2DG), Boulogne-Billancourt, France
| | - L'Houcine Ouafik
- Aix Marseille University, CNRS, INP, Institute of NeuroPhysiopathology, Marseille, France.,APHM, CHU Nord, Service de Transfert d'Oncologie Biologique, Marseille, France
| |
Collapse
|
19
|
Takamatsu Y, Ho G, Wada R, Inoue S, Hashimoto M. Adiponectin paradox as a therapeutic target of the cancer evolvability in aging. Neoplasia 2021; 23:112-117. [PMID: 33310207 PMCID: PMC7726259 DOI: 10.1016/j.neo.2020.11.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 11/16/2020] [Accepted: 11/16/2020] [Indexed: 02/07/2023]
Abstract
Recent study suggests that protofibril-formation of amyloidogenic proteins (APs) might be involved in evolvability, an epigenetic inheritance of multiple stresses, in various biological systems. In cancer, evolvability of multiple APs, such as p53, γ-synuclein and the members of the calcitonin family of peptides, might be involved in various features, including increased cell proliferation, metastasis and medical treatment resistance. In this context, the objective of this paper is to explore the potential therapeutic benefits of reduced APs evolvability against cancer. Notably, the same APs are involved in the pathogenesis of neurodegenerative disease and cancer. Given the unsatisfactory outcomes of recent clinical trial of Aβ immunotherapy in Alzheimer's disease, it is possible that suppressing the aggregation of individual APs might also be not effective in cancer. As such, we highlight the adiponectin (APN) paradox that might be positioned upstream of AP aggregation in both neurodegenerative disease and cancer, as a common therapeutic target in both disease types. Provided that the APN paradox due to APN resistance under the diabetic conditions might promote AP aggregation, suppressing the APN paradox combined with antidiabetic treatments might be effective for the therapy of both neurodegenerative disease and cancer.
Collapse
Affiliation(s)
- Yoshiki Takamatsu
- Laboratory for Parkinson's disease, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo, Japan
| | - Gilbert Ho
- PCND Neuroscience Research Institute, Poway, CA, USA
| | - Ryoko Wada
- Laboratory for Parkinson's disease, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo, Japan
| | - Satoshi Inoue
- Department of Systems Aging Science and Medicine, Tokyo Metropolitan Institute of Gerontology, Itabashi-ku, Tokyo, Japan; Research Center for Genomic Medicine, Saitama Medical University, Saitama, Japan
| | - Makoto Hashimoto
- Laboratory for Parkinson's disease, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo, Japan.
| |
Collapse
|
20
|
Ballester V, Taylor WR, Slettedahl SW, Mahoney DW, Yab TC, Sinicrope FA, Boland CR, Lidgard GP, Cruz-Correa MR, Smyrk TC, Boardman LA, Ahlquist DA, Kisiel JB. Novel methylated DNA markers accurately discriminate Lynch syndrome associated colorectal neoplasia. Epigenomics 2020; 12:2173-2187. [PMID: 33350853 PMCID: PMC7923255 DOI: 10.2217/epi-2020-0132] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Aim: Acquired molecular changes in Lynch syndrome (LS) colorectal tumors have been largely unstudied. We identified methylated DNA markers (MDMs) for discrimination of colorectal neoplasia in LS and determined if these MDMs were comparably discriminant in sporadic patients. Patients & methods: For LS discovery, we evaluated DNA from 53 colorectal case and control tissues using next generation sequencing. For validation, blinded methylation-specific PCR assays to the selected MDMs were performed on 197 cases and controls. Results: OPLAH was the most discriminant MDM with areas under the receiver operating characteristic curve ≥0.97 for colorectal neoplasia in LS and sporadic tissues. ALKBH5, was uniquely hypermethylated in LS neoplasms. Conclusion: Highly discriminant MDMs for colorectal neoplasia in LS were identified with potential use in screening and surveillance.
Collapse
Affiliation(s)
- Veroushka Ballester
- Division of Digestive & Liver Diseases, Columbia University, New York, NY 10032, USA
| | - William R Taylor
- Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, MN 55905, USA
| | | | | | - Tracy C Yab
- Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, MN 55905, USA
| | - Frank A Sinicrope
- Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, MN 55905, USA
| | | | | | - Marcia R Cruz-Correa
- Comprehensive Cancer Center, University of Puerto Rico Medical Sciences Campus, San Juan, PR 00936, USA
| | - Thomas C Smyrk
- Department of Laboratory Medicine & Pathology, Mayo Clinic, Rochester, MN 55905, USA
| | - Lisa A Boardman
- Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, MN 55905, USA
| | - David A Ahlquist
- Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, MN 55905, USA
| | - John B Kisiel
- Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
21
|
Paré M, Darini CY, Yao X, Chignon-Sicard B, Rekima S, Lachambre S, Virolle V, Aguilar-Mahecha A, Basik M, Dani C, Ladoux A. Breast cancer mammospheres secrete Adrenomedullin to induce lipolysis and browning of adjacent adipocytes. BMC Cancer 2020; 20:784. [PMID: 32819314 PMCID: PMC7441622 DOI: 10.1186/s12885-020-07273-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 08/09/2020] [Indexed: 01/16/2023] Open
Abstract
Background Cancer cells cooperate with cells that compose their environment to promote tumor growth and invasion. Among them, adipocytes provide lipids used as a source of energy by cancer cells and adipokines that contribute to tumor expansion. Mechanisms supporting the dynamic interactions between cancer cells and stromal adipocytes, however, remain unclear. Methods We set-up a co-culture model with breast cancer cells grown in 3D as mammospheres and human adipocytes to accurately recapitulate intrinsic features of tumors, such as hypoxia and cancer cell–adipocytes interactions. Results Herein, we observed that the lipid droplets’ size was reduced in adipocytes adjacent to the mammospheres, mimicking adipocyte morphology on histological sections. We showed that the uncoupling protein UCP1 was expressed in adipocytes close to tumor cells on breast cancer histological sections as well as in adipocytes in contact with the mammospheres. Mammospheres produced adrenomedullin (ADM), a multifactorial hypoxia-inducible peptide while ADM receptors were detected in adipocytes. Stimulation of adipocytes with ADM promoted UCP1 expression and increased HSL phosphorylation, which activated lipolysis. Invalidation of ADM in breast cancer cells dramatically reduced UCP1 expression in adipocytes. Conclusions Breast tumor cells secreted ADM that modified cancer–associated adipocytes through paracrine signaling, leading to metabolic changes and delipidation. Hence, ADM appears to be crucial in controlling the interactions between cancer cells and adipocytes and represents an excellent target to hinder them.
Collapse
Affiliation(s)
- Martin Paré
- Université Côte d'Azur, CNRS, INSERM, iBV, Nice, France
| | - Cédric Y Darini
- Segal Cancer Center, Lady Davis Institute for Medical Research, Sir Mortimer B. Davis Jewish General Hospital, McGill University, Montréal, Québec, Canada
| | - Xi Yao
- Université Côte d'Azur, CNRS, INSERM, iBV, Nice, France
| | - Bérengère Chignon-Sicard
- Université Côte d'Azur, Pasteur 2 Hospital, Department of Plastic and Reconstructive Surgery, Nice, France
| | - Samah Rekima
- Université Côte d'Azur, CNRS, INSERM, iBV, Nice, France
| | | | | | - Adriana Aguilar-Mahecha
- Segal Cancer Center, Lady Davis Institute for Medical Research, Sir Mortimer B. Davis Jewish General Hospital, McGill University, Montréal, Québec, Canada
| | - Mark Basik
- Segal Cancer Center, Lady Davis Institute for Medical Research, Sir Mortimer B. Davis Jewish General Hospital, McGill University, Montréal, Québec, Canada.,Division of Experimental Medicine, McGill University, Montréal, Québec, Canada
| | | | - Annie Ladoux
- Université Côte d'Azur, CNRS, INSERM, iBV, Nice, France.
| |
Collapse
|
22
|
Avgoustou P, Jailani ABA, Zirimwabagabo JO, Tozer MJ, Gibson KR, Glossop PA, Mills JEJ, Porter RA, Blaney P, Bungay PJ, Wang N, Shaw AP, Bigos KJA, Holmes JL, Warrington JI, Skerry TM, Harrity JPA, Richards GO. Discovery of a First-in-Class Potent Small Molecule Antagonist against the Adrenomedullin-2 Receptor. ACS Pharmacol Transl Sci 2020; 3:706-719. [PMID: 32832872 DOI: 10.1021/acsptsci.0c00032] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Indexed: 01/10/2023]
Abstract
The hormone adrenomedullin has both physiological and pathological roles in biology. As a potent vasodilator, adrenomedullin is critically important in the regulation of blood pressure, but it also has several roles in disease, of which its actions in cancer are becoming recognized to have clinical importance. Reduced circulating adrenomedullin causes increased blood pressure but also reduces tumor progression, so drugs blocking all effects of adrenomedullin would be unacceptable clinically. However, there are two distinct receptors for adrenomedullin, each comprising the same G protein-coupled receptor (GPCR), the calcitonin receptor-like receptor (CLR), together with a different accessory protein known as a receptor activity-modifying protein (RAMP). The CLR with RAMP2 forms an adrenomedullin-1 receptor, and the CLR with RAMP3 forms an adrenomedullin-2 receptor. Recent research suggests that a selective blockade of adrenomedullin-2 receptors would be therapeutically valuable. Here we describe the design, synthesis, and characterization of potent small-molecule adrenomedullin-2 receptor antagonists with 1000-fold selectivity over the adrenomedullin-1 receptor, although retaining activity against the CGRP receptor. These molecules have clear effects on markers of pancreatic cancer progression in vitro, drug-like pharmacokinetic properties, and inhibit xenograft tumor growth and extend life in a mouse model of pancreatic cancer. Taken together, our data support the promise of a new class of anticancer therapeutics as well as improved understanding of the pharmacology of the adrenomedullin receptors and other GPCR/RAMP heteromers.
Collapse
Affiliation(s)
- Paris Avgoustou
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, S10 2TN, U.K
| | - Ameera B A Jailani
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, S10 2TN, U.K
| | | | | | - Karl R Gibson
- Sandexis Medicinal Chemistry Ltd, Sandwich, Kent CT13 9ND, U.K
| | - Paul A Glossop
- Sandexis Medicinal Chemistry Ltd, Sandwich, Kent CT13 9ND, U.K
| | - James E J Mills
- Sandexis Medicinal Chemistry Ltd, Sandwich, Kent CT13 9ND, U.K
| | | | - Paul Blaney
- Concept Life Sciences, High Peak, SK23 0PG, U.K
| | - Peter J Bungay
- Sympetrus Ltd., Bishop's Stortford, Hertfordshire CM23 3BT, U.K
| | - Ning Wang
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, S10 2TN, U.K
| | - Alice P Shaw
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, S10 2TN, U.K
| | - Kamilla J A Bigos
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, S10 2TN, U.K
| | - Joseph L Holmes
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, S10 2TN, U.K
| | - Jessica I Warrington
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, S10 2TN, U.K
| | - Timothy M Skerry
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, S10 2TN, U.K
| | - Joseph P A Harrity
- Department of Chemistry, University of Sheffield, Sheffield, S10 2TN, U.K
| | - Gareth O Richards
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, S10 2TN, U.K
| |
Collapse
|
23
|
Tian Z, Wang Z, Chen Y, Qu S, Liu C, Chen F, Ma L, Zhu J. Bioinformatics Analysis of Prognostic Tumor Microenvironment-Related Genes in the Tumor Microenvironment of Hepatocellular Carcinoma. Med Sci Monit 2020; 26:e922159. [PMID: 32231177 PMCID: PMC7146066 DOI: 10.12659/msm.922159] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Background Growing evidence shows that the tumor microenvironment plays a crucial role in the pathogenesis of hepatocellular carcinoma (HCC). The present work aimed to screen tumor microenvironment-related genes strongly related to prognosis and to construct a prognostic gene expression model for HCC. Material/Methods We downloaded gene expression data of 371 HCC patients in The Cancer Genome Atlas (TCGA). A novel ESTIMATE algorithm was applied to calculate immune scores and stromal scores for each patient. Then, the differentially-expressed genes (DEGs) were detected according to the immune and stromal scores, and tumor microenvironment-related genes were further explored. Univariate, Lasso, and multivariate Cox analyses were performed to build the tumor microenvironment-related prediction model. Results Stromal and immune scores were calculated and were found to be correlated with the 3-year prognosis of HCC patients. DEGs were detected according to the stromal and immune scores. There were 49 genes with prognostic value in both TCGA and ICGC (International Cancer Genome Consortium) considered as prognostic tumor microenvironment-related genes. Univariate, Lasso, and multivariate Cox analyses were conducted. A novel 2-gene signature (IL18RAP and GPR182) was built for HCC 3-year prognosis prediction. The 2-gene signature was regarded as an independent prognostic predictor that was correlated with 3-year survival rate, as shown by Cox regression analysis. Conclusions This study offers a novel 2-gene signature to predict overall survival of patients with HCC, which has the potential to be used as an independent prognostic predictor. Overall, this study reveals more details about the tumor microenvironment in HCC and offers novel candidate biomarkers.
Collapse
Affiliation(s)
- Zongbiao Tian
- Department of Gastroenterology, TengZhou Central People's Hospital, Tengzhou, Shandong, China (mainland)
| | - Zheng Wang
- Department of Infectious Diseases, Qilu Hospital, Shandong University, Jinan, Shandong, China (mainland)
| | - Yanfeng Chen
- Department of Gastroenterology, TengZhou Central People's Hospital, Tengzhou, Shandong, China (mainland)
| | - Shuoying Qu
- Department of Clinical Laboratory, Qilu Hospital, Shandong University, Jinan, Shandong, China (mainland)
| | - Changhong Liu
- Department of Gastroenterology, Shandong Provincial Qianfoshan Hospital, The First Hospital Affiliated with Shandong First Medical University, Jinan, Shandong, China (mainland)
| | - Fengzhe Chen
- Department of Infectious Diseases, Qilu Hospital, Shandong University, Jinan, Shandong, China (mainland)
| | - Lixian Ma
- Department of Infectious Diseases, Qilu Hospital, Shandong University, Jinan, Shandong, China (mainland)
| | - Jie Zhu
- Department of Infectious Diseases, Qilu Hospital, Shandong University, Jinan, Shandong, China (mainland)
| |
Collapse
|
24
|
Deficiency of the adrenomedullin-RAMP3 system suppresses metastasis through the modification of cancer-associated fibroblasts. Oncogene 2019; 39:1914-1930. [PMID: 31754214 DOI: 10.1038/s41388-019-1112-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 11/06/2019] [Accepted: 11/08/2019] [Indexed: 02/07/2023]
Abstract
Tumor metastasis is a primary source of morbidity and mortality in cancer. Adrenomedullin (AM) is a multifunctional peptide regulated by receptor activity-modifying proteins (RAMPs). We previously reported that the AM-RAMP2 system is involved in tumor angiogenesis, but the function of the AM-RAMP3 system remains largely unknown. Here, we investigated the actions of the AM-RAMP2 and 3 systems in the tumor microenvironment and their impact on metastasis. PAN02 pancreatic cancer cells were injected into the spleens of mice, leading to spontaneous liver metastasis. Tumor metastasis was enhanced in vascular endothelial cell-specific RAMP2 knockout mice (DI-E-RAMP2-/-). By contrast, metastasis was suppressed in RAMP3-/- mice, where the number of podoplanin (PDPN)-positive cancer-associated fibroblasts (CAFs) was reduced in the periphery of tumors at metastatic sites. Because PDPN-positive CAFs are a hallmark of tumor malignancy, we assessed the regulation of PDPN and found that Src/Cas/PDPN signaling is mediated by RAMP3. In fact, RAMP3 deficiency CAFs suppressed migration, proliferation, and metastasis in co-cultures with tumor cells in vitro and in vivo. Moreover, the activation of RAMP2 in RAMP3-/- mice suppressed both tumor growth and metastasis. Based on these results, we suggest that the upregulation of PDPN in DI-E-RAMP2-/- mice increases malignancy, while the downregulation of PDPN in RAMP3-/- mice reduces it. Selective activation of RAMP2 and inhibition of RAMP3 would therefore be expected to suppress tumor metastasis. This study provides the first evidence that understanding and targeting to AM-RAMP systems could contribute to the development of novel therapeutics against metastasis.
Collapse
|
25
|
Bhattamisra SK, Siang TC, Rong CY, Annan NC, Sean EHY, Xi LW, Lyn OS, Shan LH, Choudhury H, Pandey M, Gorain B. Type-3c Diabetes Mellitus, Diabetes of Exocrine Pancreas - An Update. Curr Diabetes Rev 2019; 15:382-394. [PMID: 30648511 DOI: 10.2174/1573399815666190115145702] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 11/02/2018] [Accepted: 01/08/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND The incidence of diabetes is increasing steeply; the number of diabetics has doubled over the past three decades. Surprisingly, the knowledge of type 3c diabetes mellitus (T3cDM) is still unclear to the researchers, scientist and medical practitioners, leading towards erroneous diagnosis, which is sometimes misdiagnosed as type 1 diabetes mellitus (T1DM), or more frequently type 2 diabetes mellitus (T2DM). This review is aimed to outline recent information on the etiology, pathophysiology, diagnostic procedures, and therapeutic management of T3cDM patients. METHODS The literature related to T3cDM was thoroughly searched from the public domains and reviewed extensively to construct this article. Further, existing literature related to the other forms of diabetes is reviewed for projecting the differences among the different forms of diabetes. Detailed and updated information related to epidemiological evidence, risk factors, symptoms, diagnosis, pathogenesis and management is structured in this review. RESULTS T3cDM is often misdiagnosed as T2DM due to the insufficient knowledge differentiating between T2DM and T3cDM. The pathogenesis of T3cDM is explained which is often linked to the history of chronic pancreatitis, pancreatic cancer. Inflammation, and fibrosis in pancreatic tissue lead to damage both endocrine and exocrine functions, thus leading to insulin/glucagon insufficiency and pancreatic enzyme deficiency. CONCLUSION Future advancements should be accompanied by the establishment of a quick diagnostic tool through the understanding of potential biomarkers of the disease and newer treatments for better control of the diseased condition.
Collapse
Affiliation(s)
- Subrat Kumar Bhattamisra
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil 57000, Kuala Lumpur, Malaysia
| | - Tiew Chin Siang
- School of Pharmacy, International Medical University, Bukit Jalil 57000, Kuala Lumpur, Malaysia
| | - Chieng Yi Rong
- School of Pharmacy, International Medical University, Bukit Jalil 57000, Kuala Lumpur, Malaysia
| | - Naveenya Chetty Annan
- School of Pharmacy, International Medical University, Bukit Jalil 57000, Kuala Lumpur, Malaysia
| | - Esther Ho Yung Sean
- School of Pharmacy, International Medical University, Bukit Jalil 57000, Kuala Lumpur, Malaysia
| | - Lim Wen Xi
- School of Pharmacy, International Medical University, Bukit Jalil 57000, Kuala Lumpur, Malaysia
| | - Ong Siu Lyn
- School of Pharmacy, International Medical University, Bukit Jalil 57000, Kuala Lumpur, Malaysia
| | - Liew Hui Shan
- School of Pharmacy, International Medical University, Bukit Jalil 57000, Kuala Lumpur, Malaysia
| | - Hira Choudhury
- Department of Pharmaceutical Technology, School of Pharmacy, International Medical University, Bukit Jalil 57000, Kuala Lumpur, Malaysia
| | - Manisha Pandey
- Department of Pharmaceutical Technology, School of Pharmacy, International Medical University, Bukit Jalil 57000, Kuala Lumpur, Malaysia
| | - Bapi Gorain
- School of Pharmacy, Taylor's University, 1, Jalan Taylors, 47500 Subang Jaya, Selangor, Malaysia
| |
Collapse
|
26
|
Pothuraju R, Rachagani S, Junker WM, Chaudhary S, Saraswathi V, Kaur S, Batra SK. Pancreatic cancer associated with obesity and diabetes: an alternative approach for its targeting. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:319. [PMID: 30567565 PMCID: PMC6299603 DOI: 10.1186/s13046-018-0963-4] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 11/14/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND Pancreatic cancer (PC) is among foremost causes of cancer related deaths worldwide due to generic symptoms, lack of effective screening strategies and resistance to chemo- and radiotherapies. The risk factors associated with PC include several metabolic disorders such as obesity, insulin resistance and type 2 diabetes mellitus (T2DM). Studies have shown that obesity and T2DM are associated with PC pathogenesis; however, their role in PC initiation and development remains obscure. MAIN BODY Several biochemical and physiological factors associated with obesity and/or T2DM including adipokines, inflammatory mediators, and altered microbiome are involved in PC progression and metastasis albeit by different molecular mechanisms. Deep understanding of these factors and causal relationship between factors and altered signaling pathways will facilitate deconvolution of disease complexity as well as lead to development of novel therapies. In the present review, we focuses on the interplay between adipocytokines, gut microbiota, adrenomedullin, hyaluronan, vanin and matrix metalloproteinase affected by metabolic alteration and pancreatic tumor progression. CONCLUSIONS Metabolic diseases, such as obesity and T2DM, contribute PC development through altered metabolic pathways. Delineating key players in oncogenic development in pancreas due to metabolic disorder could be a beneficial strategy to combat cancers associated with metabolic diseases in particular, PC.
Collapse
Affiliation(s)
- Ramesh Pothuraju
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Satyanarayana Rachagani
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Wade M Junker
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA.,Sanguine Diagnostics and Therapeutics, University of Nebraska Medical Center, Omaha, NE, USA
| | - Sanjib Chaudhary
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Viswanathan Saraswathi
- Department of Cellular & Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Sukhwinder Kaur
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA. .,Fred & Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA. .,Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
27
|
Zhang Z, Qin W, Sun Y. Contribution of biomarkers for pancreatic cancer-associated new-onset diabetes to pancreatic cancer screening. Pathol Res Pract 2018; 214:1923-1928. [PMID: 30477640 DOI: 10.1016/j.prp.2018.10.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 09/09/2018] [Accepted: 10/17/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND Pancreatic cancer (PaC) is one of the deadliest types of tumor, and it is regarded as a fatal disease, with a 5-year survival rate less than 10%. Most clinical diagnoses for PaC are made at an advanced stage because of the insidious onset of the disease, which leads to an extremely poor prognosis. RECENT FINDINGS The relationship between diabetes mellitus (DM) and PaC has been established by several decades of research, and the prevalence of DM in patients with PaC has been reported to be 40%, with half of the patients having developed new-onset DM within 2 years or less. Increasing evidence suggests that new-onset DM is associated with a high prevalence of PaC, and PaC resection ameliorates DM. Therefore, screening for PaC may be needed in patients with newly developed DM. PURPOSE The objective of this review was to present our current understanding of biomarkers for PaC-associated new-onset DM (PCAND), to offer a perspective on the prospects and problems of using this strategy for early screening to differentiate PCAND from new-onset type 2 DM not associated with PaC and to suggest candidate biomarkers to use for PaC screening in patients with new-onset DM. Finding sensitive and specific biomarkers to manage these patients constitutes a challenge for the research community and for public health policies.
Collapse
Affiliation(s)
- Zhenjun Zhang
- Institute of Hepatobiliary and Pancreatic Diseases, School of Medicine, Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, China
| | - Wenjie Qin
- Institute of Hepatobiliary and Pancreatic Diseases, School of Medicine, Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, China
| | - Yuling Sun
- Institute of Hepatobiliary and Pancreatic Diseases, School of Medicine, Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, China.
| |
Collapse
|
28
|
Adrenomedullin promotes the growth of pancreatic ductal adenocarcinoma through recruitment of myelomonocytic cells. Oncotarget 2018; 7:55043-55056. [PMID: 27391260 PMCID: PMC5342400 DOI: 10.18632/oncotarget.10393] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 06/17/2016] [Indexed: 12/20/2022] Open
Abstract
Stromal infiltration of myelomonocytic cells is a hallmark of pancreatic ductal adenocarcinoma (PDAC) and is related to a poor prognosis. However, the detailed mechanism for the recruitment of myelomonocytic cells to pancreatic cancer tissue remains unclear. In the present study, pancreatic cancer cells secreted high levels of adrenomedullin (ADM), and CD11b+ myelomonocytic cells expressed all components of ADM receptors, including GPR182, CRLR, RAMP2 and RAMP3. ADM enhanced the migration and invasion of myelomonocytic cells through activation of the MAPK, PI3K/Akt and eNOS signaling pathways, as well as the expression and activity of MMP-2. ADM also promoted the adhesion and trans-endothelial migration of myelomonocytic cells by increasing expression of VCAM-1 and ICAM-1 in endothelial cells. In addition, ADM induced macrophages and myeloid-derived suppressor cells (MDSCs) to express pro-tumor phenotypes. ADM knockdown in tumor-bearing mice or administration of AMA, an ADM antagonist, significantly inhibited the recruitment of myelomonocytic cells and tumor angiogenesis. Moreover, in vivo depletion of myelomonocytic cells using clodronate liposomes suppressed the progression of PDAC. These results reveal a novel function of ADM in PDAC, and suggest ADM is a promising target in the treatment of PDAC.
Collapse
|
29
|
Small molecules related to adrenomedullin reduce tumor burden in a mouse model of colitis-associated colon cancer. Sci Rep 2017; 7:17488. [PMID: 29235493 PMCID: PMC5727507 DOI: 10.1038/s41598-017-17573-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 11/28/2017] [Indexed: 02/07/2023] Open
Abstract
To investigate the contribution of adrenomedullin (AM) and its gene-related peptide, proadrenomedullin N-terminal 20 peptide (PAMP), to the progression and potential treatment of colon cancer we studied the effects of four small molecules (SM) related to AM and PAMP on a mouse model of colon cancer. For each SM, four experimental groups of male mice were used: (i) Control group; (ii) SM group; (iii) DSS group (injected with azoxymethane [AOM] and drank dextran sulfate sodium [DSS]); and (iv) DSS + SM group (treated with AOM, DSS, and the SM). None of the mice in groups i and ii developed tumors, whereas all mice in groups iii and iv developed colon neoplasias. No significant differences were found among mice treated with PAMP modulators (87877 and 106221). Mice that received the AM negative modulator, 16311, had worse colitis symptoms than their control counterparts, whereas mice injected with the AM positive modulator, 145425, had a lower number of tumors than their controls. SM 145425 regulated the expression of proliferation marker Lgr5 and had an impact on microbiota, preventing the DSS-elicited increase of the Bacteroides/Prevotella ratio. These results suggest that treatment with AM or with positive modulator SMs may represent a novel strategy for colon cancer.
Collapse
|
30
|
Benyahia Z, Dussault N, Cayol M, Sigaud R, Berenguer-Daizé C, Delfino C, Tounsi A, Garcia S, Martin PM, Mabrouk K, Ouafik L. Stromal fibroblasts present in breast carcinomas promote tumor growth and angiogenesis through adrenomedullin secretion. Oncotarget 2017; 8:15744-15762. [PMID: 28178651 PMCID: PMC5362520 DOI: 10.18632/oncotarget.14999] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 01/03/2017] [Indexed: 11/25/2022] Open
Abstract
Tumor- or cancer-associated fibroblasts (TAFs or CAFs) are active players in tumorigenesis and exhibit distinct angiogenic and tumorigenic properties. Adrenomedullin (AM), a multifunctional peptide plays an important role in angiogenesis and tumor growth through its receptors calcitonin receptor-like receptor/receptor activity modifying protein-2 and -3 (CLR/RAMP2 and CLR/RAMP3). We show that AM and AM receptors mRNAs are highly expressed in CAFs prepared from invasive breast carcinoma when compared to normal fibroblasts. Immunostaining demonstrates the presence of immunoreactive AM and AM receptors in the CAFs (n = 9). The proliferation of CAFs is decreased by anti-AM antibody (αAM) and anti-AM receptors antibody (αAMR) treatment, suggesting that AM may function as a potent autocrine/paracrine growth factor. Systemic administration of αAMR reduced neovascularization of in vivo Matrigel plugs containing CAFs as demonstrated by reduced numbers of the vessel structures, suggesting that AM is one of the CAFs-derived factors responsible for endothelial cell-like and pericytes recruitment to built a neovascularization. We show that MCF-7 admixed with CAFs generated tumors of greater volume significantly different from the MCF-7 xenografts in nude mice due in part to the induced angiogenesis. αAMR and AM22-52 therapies significantly suppressed the growth of CAFs/MCF-7 tumors. Histological examination of tumors treated with AM22-52 and aAMR showed evidence of disruption of tumor vasculature with depletion of vascular endothelial cells, induced apoptosis and decrease of tumor cell proliferation. Our findings highlight the importance of CAFs-derived AM pathway in growth of breast carcinoma and in neovascularization by supplying and amplifying signals that are essential for pathologic angiogenesis.
Collapse
Affiliation(s)
- Zohra Benyahia
- Aix Marseille University, The Institut National pour la Recherche Médicale, Centre de Recherche en Oncologie et Oncopharmacologie, UMR 911, 13005, Marseille, France
| | - Nadège Dussault
- Aix Marseille University, The Institut National pour la Recherche Médicale, Centre de Recherche en Oncologie et Oncopharmacologie, UMR 911, 13005, Marseille, France
| | - Mylène Cayol
- Aix Marseille University, The Institut National pour la Recherche Médicale, Centre de Recherche en Oncologie et Oncopharmacologie, UMR 911, 13005, Marseille, France
| | - Romain Sigaud
- Aix Marseille University, The Institut National pour la Recherche Médicale, Centre de Recherche en Oncologie et Oncopharmacologie, UMR 911, 13005, Marseille, France
| | - Caroline Berenguer-Daizé
- Aix Marseille University, The Institut National pour la Recherche Médicale, Centre de Recherche en Oncologie et Oncopharmacologie, UMR 911, 13005, Marseille, France
| | - Christine Delfino
- Aix Marseille University, The Institut National pour la Recherche Médicale, Centre de Recherche en Oncologie et Oncopharmacologie, UMR 911, 13005, Marseille, France
| | - Asma Tounsi
- Aix Marseille University, The Institut National pour la Recherche Médicale, Centre de Recherche en Oncologie et Oncopharmacologie, UMR 911, 13005, Marseille, France
| | - Stéphane Garcia
- Assistance Publique Hôpitaux de Marseille, Laboratoire d'Anatomie Pathologique, 13015, Marseille, France
| | - Pierre-Marie Martin
- Aix Marseille University, The Institut National pour la Recherche Médicale, Centre de Recherche en Oncologie et Oncopharmacologie, UMR 911, 13005, Marseille, France
| | - Kamel Mabrouk
- Aix Marseille University, CNRS, ICR, UMR 7273 CROPS, 13397, Marseille, France
| | - L'Houcine Ouafik
- Aix Marseille University, The Institut National pour la Recherche Médicale, Centre de Recherche en Oncologie et Oncopharmacologie, UMR 911, 13005, Marseille, France.,Assistance Publique Hôpitaux de Marseille, Service de Transfert d'Oncologie Biologique, 13015, Marseille, France
| |
Collapse
|
31
|
Yang JC, Zhang Y, He SJ, Li MM, Cai XL, Wang H, Xu LM, Cao J. TM4SF1 Promotes Metastasis of Pancreatic Cancer via Regulating the Expression of DDR1. Sci Rep 2017; 7:45895. [PMID: 28368050 PMCID: PMC5377454 DOI: 10.1038/srep45895] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 03/07/2017] [Indexed: 12/30/2022] Open
Abstract
Transmembrane-4-L-six-family-1(TM4SF1), a four-transmembrane L6 family member, is highly expressed in various pancreatic cancer cell lines and promotes cancer cells metastasis. However, the TM4SF1-associated signaling network in metastasis remains unknown. In the present study, we found that TM4SF1 affected the formation and function of invadopodia. Silencing of TM4SF1 reduced the expression of DDR1 significantly in PANC-1 and AsPC-1 cells. Through double fluorescence immuno-staining and Co-immunoprecipitation, we also found that TM4SF1 colocalized with DDR1 and had an interaction with DDR1. In addition, upregulating the expression of DDR1 rescued the inhibitory effects of cell migration and invasion, the expression of MMP2 and MMP9 and the formation and function of invadopodia when TM4SF1 silenced. In pancreatic cancer tissues, qRT-PCR and scatter plots analysis further determined that TM4SF1 had a correlation with DDR1. Collectively, our study provides a novel regulatory pathway involving TM4SF1, DDR1, MMP2 and MMP9, which promotes the formation and function of invadopodia to support cell migration and invasion in pancreatic cancer.
Collapse
Affiliation(s)
- Jia-Chun Yang
- Department of Gastroenterology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Yi Zhang
- Department of Gastroenterology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Si-Jia He
- Department of Gastroenterology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Ming-Ming Li
- Department of Gastroenterology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Xiao-Lei Cai
- Department of Gastroenterology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Hui Wang
- Department of Gastroenterology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Lei-Ming Xu
- Department of Gastroenterology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Jia Cao
- Department of Gastroenterology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, 1665 Kongjiang Road, Shanghai, 200092, China
| |
Collapse
|
32
|
Kechele DO, Blue RE, Zwarycz B, Espenschied ST, Mah AT, Siegel MB, Perou CM, Ding S, Magness ST, Lund PK, Caron KM. Orphan Gpr182 suppresses ERK-mediated intestinal proliferation during regeneration and adenoma formation. J Clin Invest 2017; 127:593-607. [PMID: 28094771 DOI: 10.1172/jci87588] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 11/22/2016] [Indexed: 12/16/2022] Open
Abstract
Orphan GPCRs provide an opportunity to identify potential pharmacological targets, yet their expression patterns and physiological functions remain challenging to elucidate. Here, we have used a genetically engineered knockin reporter mouse to map the expression pattern of the Gpr182 during development and adulthood. We observed that Gpr182 is expressed at the crypt base throughout the small intestine, where it is enriched in crypt base columnar stem cells, one of the most active stem cell populations in the body. Gpr182 knockdown had no effect on homeostatic intestinal proliferation in vivo, but led to marked increases in proliferation during intestinal regeneration following irradiation-induced injury. In the ApcMin mouse model, which forms spontaneous intestinal adenomas, reductions in Gpr182 led to more adenomas and decreased survival. Loss of Gpr182 enhanced organoid growth efficiency ex vivo in an EGF-dependent manner. Gpr182 reduction led to increased activation of ERK1/2 in basal and challenge models, demonstrating a potential role for this orphan GPCR in regulating the proliferative capacity of the intestine. Importantly, GPR182 expression was profoundly reduced in numerous human carcinomas, including colon adenocarcinoma. Together, these results implicate Gpr182 as a negative regulator of intestinal MAPK signaling-induced proliferation, particularly during regeneration and adenoma formation.
Collapse
|
33
|
Adrenomedullin promotes angiogenesis in epithelial ovarian cancer through upregulating hypoxia-inducible factor-1α and vascular endothelial growth factor. Sci Rep 2017; 7:40524. [PMID: 28091613 PMCID: PMC5238385 DOI: 10.1038/srep40524] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 12/07/2016] [Indexed: 01/18/2023] Open
Abstract
Adrenomedullin (ADM) is a multi-functional peptide related to many kinds of tumors. This study was aimed to investigate the role of ADM on angiogenesis in epithelial ovarian cancer (EOC) and its possible mechanism. The expressions of ADM, vascular endothelial growth factor (VEGF), hypoxia-inducible factor-1α (HIF-1α) and CD34 were examined by immunohistochemistry staining. The relationship among ADM, HIF-1α, VEGF and micro-vessel density (MVD) was assessed in 56 EOC tissues. CAOV3 cells were stably transfected with pcDNA-ADM (plasmid overexpressing ADM gene) or pRNA-shADM (small interfering RNA for ADM gene). Real-time PCR and western blot analysis were performed to detect the expressions of HIF-1α and VEGF. The MTT, transwell migration assay and in vitro tube formation analysis were used to evaluate the proliferation, migration, and tube formation ability of human umbilical vein endothelial cells (HUVECs) which were pretreated with ADM or ADM receptor antagonist ADM22-52. Our findings showed that ADM expression was positively correlated with the expressions of HIF-1α, VEGF or MVD in EOC. ADM upregulated expression of HIF-1α and VEGF in CAOV3 cells. ADM promoted HUVECs proliferation, migration and tube formation. In conclusion, ADM was an upstream molecule of HIF-1α/VEGF and it promoted angiogenesis through upregulating HIF-1α/VEGF in EOC.
Collapse
|
34
|
Li H, Zhu J, Chen S, Jia L, Ma Y. Fabrication of aqueous-based dual drug loaded silk fibroin electrospun nanofibers embedded with curcumin-loaded RSF nanospheres for drugs controlled release. RSC Adv 2017. [DOI: 10.1039/c7ra12394a] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
This paper presents a new nanofabrication method for dual drug loaded regenerated silk fibroin (RSF) nanofibers, based on a simple, colloid-electrospinning technique.
Collapse
Affiliation(s)
- Huijun Li
- College of Materials Science and Engineering
- Taiyuan University of Technology
- Taiyuan
- P. R. China
| | - Jingxin Zhu
- College of Materials Science and Engineering
- Taiyuan University of Technology
- Taiyuan
- P. R. China
| | - Song Chen
- College of Materials Science and Engineering
- Taiyuan University of Technology
- Taiyuan
- P. R. China
| | - Lan Jia
- College of Materials Science and Engineering
- Taiyuan University of Technology
- Taiyuan
- P. R. China
| | - Yanlong Ma
- College of Materials Science and Engineering
- Taiyuan University of Technology
- Taiyuan
- P. R. China
| |
Collapse
|
35
|
Greillier L, Tounsi A, Berenguer-Daizé C, Dussault N, Delfino C, Benyahia Z, Cayol M, Mabrouk K, Garcia S, Martin PM, Barlesi F, Ouafik L. Functional Analysis of the Adrenomedullin Pathway in Malignant Pleural Mesothelioma. J Thorac Oncol 2016; 11:94-107. [PMID: 26762744 DOI: 10.1016/j.jtho.2015.09.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Revised: 09/25/2015] [Accepted: 09/28/2015] [Indexed: 11/18/2022]
Abstract
INTRODUCTION Malignant pleural mesothelioma (MPM) grows aggressively within the thoracic cavity and has a very low cure rate, thus highlighting the need for identification of new therapeutic targets. Adrenomedullin (AM) is a multifunctional peptide that is highly expressed in several tumors and plays an important role in angiogenesis and tumor growth after binding to its receptors, calcitonin receptor-like receptor/receptor activity-modifying protein 2 (CLR/RAMP2) and calcitonin receptor-like receptor/receptor activity-modifying protein 3 (CLR/RAMP3). METHODS Real time quantitative reverse transcriptase polymerase chain reaction (RT-PCR) was used to assess the steady-state levels of AM, CLR, RAMP2 and RAMP3 messenger RNA (mRNA) transcripts in normal pleural tissue (n=5) and MPM (n=24). The expression of these candidates at protein level was revealed by immunohistochemistry. We also characterized the expression and regulation by hypoxia of AM system in MPM cell lines and MeT-5A cells. In vitro and in vivo studies were performed to determine the functional role of AM system in MPM. RESULTS In this study, real-time quantitative reverse transcriptase polymerase chain reaction showed twofold to 10-fold higher levels of AM messenger RNA in MPM tissue than in normal pleural tissue. The MPM cell lines H2452, H2052, and human mesothelioma cell line MSTO-211H showed a significant increase in expression of AM messenger RNA under hypoxic conditions. Our results also show that AM stimulates cell proliferation in vitro through the Raf1 proto-oncogene, serine/threonine kinase (CRAF)/ Mitogen-activated protein kinase kinase 1 (MEK)/Extracellular regulated MAPKinase (ERK) pathway. Furthermore, the proliferation, migration, and invasion of MPM cells were decreased after treatment with anti-AM (αAM) and anti-AM receptor antibodies, thus indicating that MPM cells are regulated by AM. The action of AM was specific and mediated by CLR/RAMP2 and CLR/RAMP3 receptors. In vivo, αAM and AM22-52 antagonist therapies blocked angiogenesis and induced apoptosis in MSTO-211H xenografts, thereby resulting in tumor regression. Histologic examination of tumors treated with AM22-52 and αAM antibody showed evidence of disruption of tumor vasculature with depletion of vascular endothelial cells and a significant decrease in lymphatic endothelial cells. CONCLUSIONS Our findings highlight the importance of the AM pathway in growth of MPM and in neovascularization by supplying and amplifying signals that are essential for pathologic neoangiogenesis and lymphangiogenesis.
Collapse
MESH Headings
- Adrenomedullin/genetics
- Adrenomedullin/metabolism
- Animals
- Apoptosis
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Blotting, Western
- Calcitonin Receptor-Like Protein/genetics
- Calcitonin Receptor-Like Protein/metabolism
- Cell Movement
- Cell Proliferation
- Flow Cytometry
- Gene Expression Regulation, Neoplastic
- Humans
- Immunoenzyme Techniques
- Lung Neoplasms/genetics
- Lung Neoplasms/metabolism
- Lung Neoplasms/pathology
- Mesothelioma/genetics
- Mesothelioma/metabolism
- Mesothelioma/pathology
- Mesothelioma, Malignant
- Mice
- Mice, Nude
- Neovascularization, Pathologic
- Pleural Neoplasms/genetics
- Pleural Neoplasms/metabolism
- Pleural Neoplasms/pathology
- Proto-Oncogene Mas
- RNA, Messenger/genetics
- Real-Time Polymerase Chain Reaction
- Receptor Activity-Modifying Protein 2/genetics
- Receptor Activity-Modifying Protein 2/metabolism
- Receptor Activity-Modifying Protein 3/genetics
- Receptor Activity-Modifying Protein 3/metabolism
- Receptors, Adrenomedullin/genetics
- Receptors, Adrenomedullin/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Tumor Cells, Cultured
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Laurent Greillier
- Aix-Marseille University, CRO2 UMR 911, Marseille, France; INSERM, CRO2 UMR 911, Marseille, France; Assistance Publique Hopitaux de Marseille, Service d'Oncologie Multidisciplinaire et Innovations Thérapeutiques, Marseille, France.
| | - Asma Tounsi
- Aix-Marseille University, CRO2 UMR 911, Marseille, France; INSERM, CRO2 UMR 911, Marseille, France
| | - Caroline Berenguer-Daizé
- Aix-Marseille University, CRO2 UMR 911, Marseille, France; INSERM, CRO2 UMR 911, Marseille, France
| | - Nadège Dussault
- Aix-Marseille University, CRO2 UMR 911, Marseille, France; INSERM, CRO2 UMR 911, Marseille, France
| | - Christine Delfino
- Aix-Marseille University, CRO2 UMR 911, Marseille, France; INSERM, CRO2 UMR 911, Marseille, France
| | - Zohra Benyahia
- Aix-Marseille University, CRO2 UMR 911, Marseille, France; INSERM, CRO2 UMR 911, Marseille, France
| | - Mylène Cayol
- Aix-Marseille University, CRO2 UMR 911, Marseille, France; INSERM, CRO2 UMR 911, Marseille, France
| | - Kamel Mabrouk
- Aix-Marseille University, LCP UMR 6264, CROPS, Marseille, France
| | - Stéphane Garcia
- Assistance Publique Hopitaux de Marseille, Service d'Anatomie et de Cytologie Pathologiques, Marseille, France
| | - Pierre-Marie Martin
- Aix-Marseille University, CRO2 UMR 911, Marseille, France; INSERM, CRO2 UMR 911, Marseille, France; Assistance Publique Hopitaux de Marseille, Service de Transfert d'Oncologie Biologique, Marseille, France
| | - Fabrice Barlesi
- Aix-Marseille University, CRO2 UMR 911, Marseille, France; INSERM, CRO2 UMR 911, Marseille, France; Assistance Publique Hopitaux de Marseille, Service d'Oncologie Multidisciplinaire et Innovations Thérapeutiques, Marseille, France
| | - L'Houcine Ouafik
- Aix-Marseille University, CRO2 UMR 911, Marseille, France; INSERM, CRO2 UMR 911, Marseille, France; Assistance Publique Hopitaux de Marseille, Service de Transfert d'Oncologie Biologique, Marseille, France
| |
Collapse
|
36
|
Hart PA, Bellin MD, Andersen DK, Bradley D, Cruz-Monserrate Z, Forsmark CE, Goodarzi MO, Habtezion A, Korc M, Kudva YC, Pandol SJ, Yadav D, Chari ST. Type 3c (pancreatogenic) diabetes mellitus secondary to chronic pancreatitis and pancreatic cancer. Lancet Gastroenterol Hepatol 2016; 1:226-237. [PMID: 28404095 DOI: 10.1016/s2468-1253(16)30106-6] [Citation(s) in RCA: 293] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 07/25/2016] [Accepted: 07/26/2016] [Indexed: 02/07/2023]
Abstract
Diabetes mellitus is a group of diseases defined by persistent hyperglycaemia. Type 2 diabetes, the most prevalent form, is characterised initially by impaired insulin sensitivity and subsequently by an inadequate compensatory insulin response. Diabetes can also develop as a direct consequence of other diseases, including diseases of the exocrine pancreas. Historically, diabetes due to diseases of the exocrine pancreas was described as pancreatogenic or pancreatogenous diabetes mellitus, but recent literature refers to it as type 3c diabetes. It is important to note that type 3c diabetes is not a single entity; it occurs because of a variety of exocrine pancreatic diseases with varying mechanisms of hyperglycaemia. The most commonly identified causes of type 3c diabetes are chronic pancreatitis, pancreatic ductal adenocarcinoma, haemochromatosis, cystic fibrosis, and previous pancreatic surgery. In this Review, we discuss the epidemiology, pathogenesis, and clinical relevance of type 3c diabetes secondary to chronic pancreatitis and pancreatic ductal adenocarcinoma, and highlight several important knowledge gaps.
Collapse
Affiliation(s)
- Phil A Hart
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| | - Melena D Bellin
- Division of Pediatric Endocrinology and Schulze Diabetes Institute, University of Minnesota Medical Center, Minneapolis, MN, USA
| | - Dana K Andersen
- Division of Digestive Diseases and Nutrition, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - David Bradley
- Division of Endocrinology, Diabetes, and Metabolism, The Ohio State University, Wexner Medical Center, Columbus, OH, USA
| | - Zobeida Cruz-Monserrate
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Christopher E Forsmark
- Division of Gastroenterology, Hepatology, and Nutrition, University of Florida, Gainesville, FL, USA
| | - Mark O Goodarzi
- Division of Endocrinology, Diabetes, and Metabolism, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Aida Habtezion
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, CA, USA
| | - Murray Korc
- Departments of Medicine, Biochemistry, and Molecular Biology, Indiana University School of Medicine, Indiana University Simon Cancer Center, Indianapolis, IN, USA; Pancreatic Cancer Signature Center, Indiana University Simon Cancer Center, Indianapolis, IN, USA
| | - Yogish C Kudva
- Division of Endocrinology and Metabolism, Mayo Clinic, Rochester, MN, USA
| | - Stephen J Pandol
- Department of Veterans Affairs, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Dhiraj Yadav
- Division of Gastroenterology, Hepatology, and Nutrition, University of Pittsburgh and UPMC Medical Center, Pittsburgh, PA, USA; Department of Medicine, University of Pittsburgh and UPMC Medical Center, Pittsburgh, PA, USA
| | - Suresh T Chari
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
37
|
Martínez-Herrero S, Larrayoz IM, Ochoa-Callejero L, Fernández LJ, Allueva A, Ochoa I, Martínez A. Prevention of Bone Loss in a Model of Postmenopausal Osteoporosis through Adrenomedullin Inhibition. Front Physiol 2016; 7:280. [PMID: 27445864 PMCID: PMC4928306 DOI: 10.3389/fphys.2016.00280] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 06/21/2016] [Indexed: 11/23/2022] Open
Abstract
Despite recent advances in the understanding and treatment options for osteoporosis, this condition remains a serious public health issue. Adrenomedullin (AM) is a regulatory peptide with reported activity on bone remodeling. To better understand this relationship we built an inducible knockout for AM. An outstanding feature of knockout mice is their heavier weight due, in part, to the presence of denser bones. The femur of knockout animals was denser, had more trabeculae, and a thicker growth plate than wild type littermates. The endocrine influence of AM on bone seems to be elicited through an indirect mechanism involving, at least, the regulation of insulin, glucose, ghrelin, and calcitonin gene-related peptide (CGRP). To confirm the data we performed a pharmacological approach using the AM inhibitor 16311 in a mouse model of osteoporosis. Ovariectomized females showed significant bone mass loss, whereas ovariectomized females treated with 16311 had similar bone density to sham operated females. In conclusion, we propose the use of AM inhibitors for the treatment of osteoporosis and other conditions leading to the loss of bone mass.
Collapse
Affiliation(s)
- Sonia Martínez-Herrero
- Angiogenesis Interest Group, Oncology Area, Center for Biomedical Research of La Rioja, Fundación Rioja Salud Logroño, Spain
| | - Ignacio M Larrayoz
- Angiogenesis Interest Group, Oncology Area, Center for Biomedical Research of La Rioja, Fundación Rioja Salud Logroño, Spain
| | - Laura Ochoa-Callejero
- Angiogenesis Interest Group, Oncology Area, Center for Biomedical Research of La Rioja, Fundación Rioja Salud Logroño, Spain
| | - Luis J Fernández
- Centro de Investigación Biomédica en Red, Aragon Institute of Health SciencesZaragoza, Spain; Group of Structural Mechanics and Materials Modelling, Aragón Institute of Engineering Research (I3A), University of ZaragozaZaragoza, Spain
| | - Alexis Allueva
- Centro de Investigación Biomédica en Red, Aragon Institute of Health SciencesZaragoza, Spain; Group of Structural Mechanics and Materials Modelling, Aragón Institute of Engineering Research (I3A), University of ZaragozaZaragoza, Spain
| | - Ignacio Ochoa
- Centro de Investigación Biomédica en Red, Aragon Institute of Health SciencesZaragoza, Spain; Group of Structural Mechanics and Materials Modelling, Aragón Institute of Engineering Research (I3A), University of ZaragozaZaragoza, Spain
| | - Alfredo Martínez
- Angiogenesis Interest Group, Oncology Area, Center for Biomedical Research of La Rioja, Fundación Rioja Salud Logroño, Spain
| |
Collapse
|
38
|
Bhardwaj A, Srivastava SK, Singh S, Tyagi N, Arora S, Carter JE, Khushman M, Singh AP. MYB Promotes Desmoplasia in Pancreatic Cancer through Direct Transcriptional Up-regulation and Cooperative Action of Sonic Hedgehog and Adrenomedullin. J Biol Chem 2016; 291:16263-70. [PMID: 27246849 DOI: 10.1074/jbc.m116.732651] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Indexed: 01/05/2023] Open
Abstract
Extensive desmoplasia is a prominent pathological characteristic of pancreatic cancer (PC) that not only impacts tumor development, but therapeutic outcome as well. Recently, we demonstrated a novel role of MYB, an oncogenic transcription factor, in PC growth and metastasis. Here we studied its effect on pancreatic tumor histopathology and associated molecular and biological mechanisms. Tumor-xenografts derived from orthotopic-inoculation of MYB-overexpressing PC cells exhibited far-greater desmoplasia in histological analyses compared with those derived from MYB-silenced PC cells. These findings were further confirmed by immunostaining of tumor-xenograft sections with collagen-I, fibronectin (major extracellular-matrix proteins), and α-SMA (well-characterized marker of myofibroblasts or activated pancreatic stellate cells (PSCs)). Likewise, MYB-overexpressing PC cells provided significantly greater growth benefit to PSCs in a co-culture system as compared with the MYB-silenced cells. Interrogation of deep-sequencing data from MYB-overexpressing versus -silenced PC cells identified Sonic-hedgehog (SHH) and Adrenomedullin (ADM) as two differentially-expressed genes among others, which encode for secretory ligands involved in tumor-stromal cross-talk. In-silico analyses predicted putative MYB-binding sites in SHH and ADM promoters, which was later confirmed by chromatin-immunoprecipitation. A cooperative role of SHH and ADM in growth promotion of PSCs was confirmed in co-culture by using their specific-inhibitors and exogenous recombinant-proteins. Importantly, while SHH acted exclusively in a paracrine fashion on PSCs and influenced the growth of PC cells only indirectly, ADM could directly impact the growth of both PC cells and PSCs. In summary, we identified MYB as novel regulator of pancreatic tumor desmoplasia, which is suggestive of its diverse roles in PC pathobiology.
Collapse
Affiliation(s)
| | | | - Seema Singh
- From the Departments of Oncologic Sciences and Departments of Biochemistry and Molecular Biology and
| | | | - Sumit Arora
- From the Departments of Oncologic Sciences and
| | - James E Carter
- Pathology, College of Medicine, University of South Alabama, Mobile, Alabama 36688
| | - Moh'd Khushman
- Interdisciplinary Clinical Oncology, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama 36604 and
| | - Ajay P Singh
- From the Departments of Oncologic Sciences and Departments of Biochemistry and Molecular Biology and
| |
Collapse
|
39
|
Khalfaoui-Bendriss G, Dussault N, Fernandez-Sauze S, Berenguer-Daizé C, Sigaud R, Delfino C, Cayol M, Metellus P, Chinot O, Mabrouk K, Martin PM, Ouafik L. Adrenomedullin blockade induces regression of tumor neovessels through interference with vascular endothelial-cadherin signalling. Oncotarget 2016; 6:7536-53. [PMID: 25924235 PMCID: PMC4480698 DOI: 10.18632/oncotarget.3167] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Accepted: 01/19/2015] [Indexed: 11/25/2022] Open
Abstract
The cellular and molecular mechanisms by which adrenomedullin (AM) blockade suppresses tumor neovessels are not well defined. Herein, we show that AM blockade using anti-AM and anti-AM receptors antibodies targets vascular endothelial cells (ECs) and vascular smooth muscle cells (VSMCs), and induces regression of unstable nascent tumor neovessels. The underlying mechanism involved, and shown in vitro and in vivo in mice, is the disruption of the molecular engagement of the endothelial cell-specific junctional molecules vascular endothelial-cadherin (VE-cadherin)/β-catenin complex. AM blockade increases endothelial cell permeability by inhibiting cell-cell contacts predominantly through disruption of VE-cadherin/β-catenin/Akt signalling pathway, thereby leading to vascular collapse and regression of tumor neovessels. At a molecular level, we show that AM blockade induces tyrosine phosphorylation of VE-cadherin at a critical tyrosine, Tyr731, which is sufficient to prevent the binding of β-catenin to the cytoplasmic tail of VE-cadherin leading to the inhibition of cell barrier function. Furthermore, we demonstrate activation of Src kinase by phosphorylation on Tyr416, supporting a role of Src to phosphorylate Tyr731-VE-cadherin. In this model, Src inhibition impairs αAM and αAMR-induced Tyr731-VE-cadherin phosphorylation in a dose-dependent manner, indicating that Tyr731-VE-cadherin phosphorylation state is dependent on Src activation. We found that AM blockade induces β-catenin phosphorylation on Ser33/Ser37/Thr41 sites in both ECs and VSMCs both in vitro and in vivo in mice. These data suggest that AM blockade selectively induces regression of unstable tumor neovessels, through disruption of VE-cadherin signalling. Targeting AM system may present a novel therapeutic target to selectively disrupt assembly and induce regression of nascent tumor neovessels, without affecting normal stabilized vasculature.
Collapse
Affiliation(s)
- Ghizlane Khalfaoui-Bendriss
- Aix Marseille Université, CRO2, UMR_S 911, Faculté de Médecine, Marseille, France.,Inserm, U911-CRO2, Marseille, France
| | - Nadège Dussault
- Aix Marseille Université, CRO2, UMR_S 911, Faculté de Médecine, Marseille, France.,Inserm, U911-CRO2, Marseille, France
| | - Samantha Fernandez-Sauze
- Aix Marseille Université, CRO2, UMR_S 911, Faculté de Médecine, Marseille, France.,Inserm, U911-CRO2, Marseille, France
| | - Caroline Berenguer-Daizé
- Aix Marseille Université, CRO2, UMR_S 911, Faculté de Médecine, Marseille, France.,Inserm, U911-CRO2, Marseille, France
| | - Romain Sigaud
- Aix Marseille Université, CRO2, UMR_S 911, Faculté de Médecine, Marseille, France.,Inserm, U911-CRO2, Marseille, France
| | - Christine Delfino
- Aix Marseille Université, CRO2, UMR_S 911, Faculté de Médecine, Marseille, France.,Inserm, U911-CRO2, Marseille, France
| | - Mylène Cayol
- Aix Marseille Université, CRO2, UMR_S 911, Faculté de Médecine, Marseille, France.,Inserm, U911-CRO2, Marseille, France
| | - Philippe Metellus
- Aix Marseille Université, CRO2, UMR_S 911, Faculté de Médecine, Marseille, France.,Inserm, U911-CRO2, Marseille, France
| | - Olivier Chinot
- Aix Marseille Université, CRO2, UMR_S 911, Faculté de Médecine, Marseille, France.,Inserm, U911-CRO2, Marseille, France
| | - Kamel Mabrouk
- Aix-Marseille Université, CNRS, UMR 7273, Institut de Chimie Radicalaire (ICR) Marseille, France
| | - Pierre-Marie Martin
- Aix Marseille Université, CRO2, UMR_S 911, Faculté de Médecine, Marseille, France.,Inserm, U911-CRO2, Marseille, France.,AP-HM, CHU Nord, Service de Transfert d'Oncologie Biologique, Marseille, France
| | - L'Houcine Ouafik
- Aix Marseille Université, CRO2, UMR_S 911, Faculté de Médecine, Marseille, France.,Inserm, U911-CRO2, Marseille, France.,AP-HM, CHU Nord, Service de Transfert d'Oncologie Biologique, Marseille, France
| |
Collapse
|
40
|
Adrenomedullin: A potential therapeutic target for retinochoroidal disease. Prog Retin Eye Res 2016; 52:112-29. [DOI: 10.1016/j.preteyeres.2016.01.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 01/06/2016] [Accepted: 01/07/2016] [Indexed: 11/22/2022]
|
41
|
Cao J, Yang J, Ramachandran V, Arumugam T, Deng D, Li Z, Xu L, Logsdon CD. TM4SF1 Promotes Gemcitabine Resistance of Pancreatic Cancer In Vitro and In Vivo. PLoS One 2015; 10:e0144969. [PMID: 26709920 PMCID: PMC4692438 DOI: 10.1371/journal.pone.0144969] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 11/25/2015] [Indexed: 11/26/2022] Open
Abstract
Background TM4SF1 is overexpressed in pancreatic ductal adenocarcinoma (PDAC) and affects the development of this cancer. Also, multidrug resistance (MDR) is generally associated with tumor chemoresistance in pancreatic cancer. However, the correlation between TM4SF1 and MDR remains unknown. This research aims to investigate the effect of TM4SF1 on gemcitabine resistance in PDAC and explore the possible molecular mechanism between TM4SF1 and MDR. Methods The expression of TM4SF1 was evaluated in pancreatic cancer cell lines and human pancreatic duct epithelial (HPDE) cell lines by quantitative RT-PCR. TM4SF1 siRNA transfection was carried out using Hiperfect transfection reagent to knock down TM4SF1. The transcripts were analyzed by quantitative RT-PCR, RT-PCR and western blotting for further study. The cell proliferation and apoptosis were obtained to investigate the sensitivity to gemcitabine of pancreatic cancer cells after silencing TM4SF1 in vitro. We demonstrated that cell signaling of TM4SF1 mediated chemoresistance in cancer cells by assessing the expression of multidrug resistance (MDR) genes using quantitative RT-PCR. In vivo, we used orthotopic pancreatic tumor models to investigate the effect of proliferation after silencing TM4SF1 by a lentivirus-mediated shRNA in MIA PaCa-2 cell lines. Results The mRNA expression of TM4SF1 was higher in seven pancreatic cancer cell lines than in HPDE cell lines. In three gemcitabine-sensitive cell lines (L3.6pl, BxPC-3, SU86.86), the expression of TM4SF1 was lower than that in four gemcitabine-resistant cell lines (MIA PaCa-2, PANC-1, Hs766T, AsPC-1). We evaluated that TM4SF1 was a putative target for gemcitabine resistance in pancreatic cancer cells. Using AsPC-1, MIA PaCa-2 and PANC-1, we investigated that TM4SF1 silencing affected cell proliferation and increased the percentages of cell apoptosis mediated by treatment with gemcitabine compared with cells which were treated with negative control. This resistance was associated with the expression of multidrug resistance genes including ABCB1 and ABCC1. In vivo, silencing of TM4SF1 in MIA PaCa-2 cell lines increased the effectiveness of gemcitabine-based treatment in orthotopic pancreatic tumor models evaluated using noninvasive bioluminescent imaging. Conclusion These findings suggest that TM4SF1 is a surface membrane antigen that is highly expressed in pancreatic cancer cells and increases the chemoresistance to gemcitabine. Thus, TM4SF1 may be a promising target to overcome the chemoresistance of pancreatic cancer.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B/biosynthesis
- Animals
- Antigens, Surface/genetics
- Apoptosis/drug effects
- Carcinoma, Pancreatic Ductal/genetics
- Carcinoma, Pancreatic Ductal/pathology
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Deoxycytidine/analogs & derivatives
- Deoxycytidine/pharmacology
- Drug Resistance, Multiple/genetics
- Drug Resistance, Neoplasm/genetics
- Humans
- Male
- Mice
- Mice, Nude
- Multidrug Resistance-Associated Proteins/biosynthesis
- Neoplasm Proteins/genetics
- Pancreatic Neoplasms/drug therapy
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/pathology
- RNA Interference
- RNA, Messenger/biosynthesis
- RNA, Small Interfering/genetics
- Signal Transduction/genetics
- Gemcitabine
Collapse
Affiliation(s)
- Jia Cao
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200092, China
| | - Jiachun Yang
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200092, China
| | - Vijaya Ramachandran
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, 77030, United States of America
| | - Thiruvengadam Arumugam
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, 77030, United States of America
| | - Defeng Deng
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, 77030, United States of America
| | - Zhaoshen Li
- Department of Gastroenterology, Changhai Hospital, the Second Military Medical University, Shanghai, 200433, China
| | - Leiming Xu
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200092, China
- * E-mail: (LMX); (CDL)
| | - Craig D. Logsdon
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, 77030, United States of America
- * E-mail: (LMX); (CDL)
| |
Collapse
|
42
|
Zhou C, Zheng Y, Li L, Zhai W, Li R, Liang Z, Zhao L. Adrenomedullin promotes intrahepatic cholangiocellular carcinoma metastasis and invasion by inducing epithelial-mesenchymal transition. Oncol Rep 2015; 34:610-6. [PMID: 26043778 PMCID: PMC4508861 DOI: 10.3892/or.2015.4034] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2015] [Accepted: 05/18/2015] [Indexed: 12/15/2022] Open
Abstract
Intrahepatic cholangiocellular carcinoma (ICC) is the second most common type of primary liver cancer. However, its etiology and molecular pathogenesis remain largely unknown. The present study aimed to investigate the association between adrenomedullin (ADM) and epithelial-mesenchymal transition (EMT) in ICC and to elucidate the underlying signaling pathway. We evaluated the clinical significance of ADM in 133 ICC patients using tissue microarray analysis of ICC tissues. We also investigated the mechanisms of ADM in ICC EMT-mediated metastasis in cholangiocarci-noma cell lines in vitro. The results revealed that ADM was upregulated in human ICC tissues (73/133) compared with that in healthy controls. ADM expression was positively correlated with shorter overall survival (P<0.01). The characteristics of EMT were induced in vitro by adenoviral transduction of ADM into HuCCT1 cells, resulting in the downregulation of E-cadherin and ZO-1, and the concomitant upregulation of N-cadherin and vimentin. Knockdown of ADM by short hairpin RNA in HUH28 cells expressing high levels of ADM was associated with the reversal of EMT. Functional studies revealed that ADM regulated the activation of ZEB1, which subsequently mediated EMT. The results of the present study suggest that ADM plays an important role in ICC metastasis, and that ADM signaling of EMT may represent a valuable therapeutic target in cancer patients.
Collapse
Affiliation(s)
- Chuang Zhou
- Department of General Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Yan Zheng
- Institutes of Biomedical Science, Fudan University, Shanghai 200032, P.R. China
| | - Lin Li
- Department of Dermatology, Zhengzhou Children's Hospital, Zhengzhou, Henan 450053, P.R. China
| | - Wenlong Zhai
- Department of General Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Renfeng Li
- Department of General Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Zhiwen Liang
- Department of General Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Longshuan Zhao
- Department of General Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
43
|
Wu XY, Hao CP, Ling M, Guo CH, Ma W. Hypoxia-induced apoptosis is blocked by adrenomedullin via upregulation of Bcl-2 in human osteosarcoma cells. Oncol Rep 2015; 34:787-94. [PMID: 26035796 DOI: 10.3892/or.2015.4011] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 05/11/2015] [Indexed: 11/06/2022] Open
Abstract
Adrenomedullin (ADM), a multifunctional regulatory peptide, is potentially induced by hypoxia in physiological and pathological tissues, including many types of malignant tumors. Recent research has demonstrated that ADM expression is highly associated with the prognosis and disease severity of human osteosarcoma. However, the effect of ADM on the apoptosis of osteosarcoma cells and its possible mechanism remain to be elucidated. In the present study, we observed that mRNA and protein levels of ADM were increased in human osteosarcoma SOSP-F5M2 cells under a hypoxic microenvironment induced by cobalt chloride (CoCl2) in a time-dependent manner. Treatment with ADM significantly blunted hypoxic-induced apoptosis, evaluated by Hoechst 33342 staining and Annexin V-FITC/PI labeling. The expression of B-cell lymphoma-2 (Bcl-2) was increased by administration of ADM; meanwhile, this effect was reversed by exogenously adding U0126, a selective inhibitor of MEK or ADM22-52 (ADM-specific receptor antagonist). These results demonstrated that ADM acted as a survival factor to inhibit hypoxic-induced apoptosis via interacting with its receptors CRLR-RAMP (2,3) in osteosarcoma cells. The anti-apoptotic function of ADM was found to be mediated by upregulation of the expression of Bcl-2 partially through activation of the MEK/ERK1/2 signaling pathway. Therefore, targeting of the ADM/ADM acceptors/ERK1/2/Bcl-2 pathway may provide a potential strategy through which to induce the apoptosis of osteosarcoma cells.
Collapse
Affiliation(s)
- Xue-Yuan Wu
- Department of Orthopaedics, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Cui-Pei Hao
- Department of Gynaecology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Ming Ling
- Department of Orthopaedics, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Chi-Hua Guo
- Department of Orthopedics, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Wei Ma
- Department of Orthopedics, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
44
|
Javeed N, Sagar G, Dutta SK, Smyrk TC, Lau JS, Bhattacharya S, Truty M, Petersen GM, Kaufman RJ, Chari ST, Mukhopadhyay D. Pancreatic Cancer-Derived Exosomes Cause Paraneoplastic β-cell Dysfunction. Clin Cancer Res 2015; 21:1722-33. [PMID: 25355928 PMCID: PMC4383684 DOI: 10.1158/1078-0432.ccr-14-2022] [Citation(s) in RCA: 139] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Accepted: 10/11/2014] [Indexed: 12/14/2022]
Abstract
PURPOSE Pancreatic cancer frequently causes diabetes. We recently proposed adrenomedullin as a candidate mediator of pancreatic β-cell dysfunction in pancreatic cancer. How pancreatic cancer-derived adrenomedullin reaches β cells remote from the cancer to induce β-cell dysfunction is unknown. We tested a novel hypothesis that pancreatic cancer sheds adrenomedullin-containing exosomes into circulation, which are transported to β cells and impair insulin secretion. EXPERIMENTAL METHODS We characterized exosomes from conditioned media of pancreatic cancer cell lines (n = 5) and portal/peripheral venous blood of patients with pancreatic cancer (n = 20). Western blot analysis showed the presence of adrenomedullin in pancreatic cancer-exosomes. We determined the effect of adrenomedullin-containing pancreatic cancer exosomes on insulin secretion from INS-1 β cells and human islets, and demonstrated the mechanism of exosome internalization into β cells. We studied the interaction between β-cell adrenomedullin receptors and adrenomedullin present in pancreatic cancer-exosomes. In addition, the effect of adrenomedullin on endoplasmic reticulum (ER) stress response genes and reactive oxygen/nitrogen species generation in β cells was shown. RESULTS Exosomes were found to be the predominant extracellular vesicles secreted by pancreatic cancer into culture media and patient plasma. Pancreatic cancer-exosomes contained adrenomedullin and CA19-9, readily entered β cells through caveolin-mediated endocytosis or macropinocytosis, and inhibited insulin secretion. Adrenomedullin in pancreatic cancer exosomes interacted with its receptor on β cells. Adrenomedullin receptor blockade abrogated the inhibitory effect of exosomes on insulin secretion. β cells exposed to adrenomedullin or pancreatic cancer exosomes showed upregulation of ER stress genes and increased reactive oxygen/nitrogen species. CONCLUSIONS Pancreatic cancer causes paraneoplastic β-cell dysfunction by shedding adrenomedullin(+)/CA19-9(+) exosomes into circulation that inhibit insulin secretion, likely through adrenomedullin-induced ER stress and failure of the unfolded protein response.
Collapse
Affiliation(s)
- Naureen Javeed
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Gunisha Sagar
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Shamit K Dutta
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Thomas C Smyrk
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Julie S Lau
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Santanu Bhattacharya
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Mark Truty
- Department of Surgery, Mayo Clinic, Rochester, Minnesota
| | - Gloria M Petersen
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | - Randal J Kaufman
- Degenerative Disease Research Program, Sanford Burnham Medical Research Institute, La Jolla, California
| | - Suresh T Chari
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota.
| | | |
Collapse
|
45
|
Arumugam T, Deng D, Bover L, Wang H, Logsdon CD, Ramachandran V. New Blocking Antibodies against Novel AGR2-C4.4A Pathway Reduce Growth and Metastasis of Pancreatic Tumors and Increase Survival in Mice. Mol Cancer Ther 2015; 14:941-51. [PMID: 25646014 PMCID: PMC4710371 DOI: 10.1158/1535-7163.mct-14-0470] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Accepted: 01/16/2015] [Indexed: 01/25/2023]
Abstract
Anterior gradient 2 (AGR2) promotes cancer growth, metastasis, and resistance to therapy via unknown mechanisms. We investigated the effects of extracellular AGR2 signaling through the orphan glycosylphosphatidylinositol-linked receptor C4.4A in pancreatic ductal adenocarcinoma (PDAC). Proliferation, migration, invasion, and apoptosis were measured using colorimetric, Boyden chamber, and FACS analyses. We developed blocking mAbs against AGR2 and C4.4A and tested their effects, along with siRNAs, on cancer cell functions and on orthotopic tumors in nude mice. Extracellular AGR2 stimulated proliferation, migration, invasion, and chemoresistance of PDAC cell lines. AGR2 interacted with C4.4A in cell lysates and mixtures of recombinant proteins. Knockdown of C4.4A reduced migration and resistance to gemcitabine. PDAC tissues, but not adjacent healthy pancreatic tissues, expressed high levels of AGR2 and C4.4A. AGR2 signaling through C4.4A required laminins 1 or 5 and integrin β1. Administration of antibodies against AGR2 and C4.4A reduced growth and metastasis and caused regression of aggressive xenograft tumors, leading to increased survival of mice. These data support a model in which AGR2 binds and signals via C4.4A in an autocrine loop and promotes the growth of pancreas tumors in mice. Blocking mAbs against AGR2 and C4.4A may have therapeutic potential against PDAC.
Collapse
Affiliation(s)
- Thiruvengadam Arumugam
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Defeng Deng
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Laura Bover
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Huamin Wang
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Craig D Logsdon
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas. Department of GI Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Vijaya Ramachandran
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
46
|
Larráyoz IM, Martínez-Herrero S, García-Sanmartín J, Ochoa-Callejero L, Martínez A. Adrenomedullin and tumour microenvironment. J Transl Med 2014; 12:339. [PMID: 25475159 PMCID: PMC4272513 DOI: 10.1186/s12967-014-0339-2] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 11/21/2014] [Indexed: 01/03/2023] Open
Abstract
Adrenomedullin (AM) is a regulatory peptide whose involvement in tumour progression is becoming more relevant with recent studies. AM is produced and secreted by the tumour cells but also by numerous stromal cells including macrophages, mast cells, endothelial cells, and vascular smooth muscle cells. Most cancer patients present high levels of circulating AM and in some cases these higher levels correlate with a worst prognosis. In some cases it has been shown that the high AM levels return to normal following surgical removal of the tumour, thus indicating the tumour as the source of this excessive production of AM. Expression of this peptide is a good investment for the tumour cell since AM acts as an autocrine/paracrine growth factor, prevents apoptosis-mediated cell death, increases tumour cell motility and metastasis, induces angiogenesis, and blocks immunosurveillance by inhibiting the immune system. In addition, AM expression gets rapidly activated by hypoxia through a HIF-1α mediated mechanism, thus characterizing AM as a major survival factor for tumour cells. Accordingly, a number of studies have shown that inhibition of this peptide or its receptors results in a significant reduction in tumour progression. In conclusion, AM is a great target for drug development and new drugs interfering with this system are being developed.
Collapse
Affiliation(s)
- Ignacio M Larráyoz
- Oncology Area, Center for Biomedical Research of La Rioja CIBIR, C/Piqueras 98, Logroño, 26006, Spain.
| | - Sonia Martínez-Herrero
- Oncology Area, Center for Biomedical Research of La Rioja CIBIR, C/Piqueras 98, Logroño, 26006, Spain.
| | - Josune García-Sanmartín
- Oncology Area, Center for Biomedical Research of La Rioja CIBIR, C/Piqueras 98, Logroño, 26006, Spain.
| | - Laura Ochoa-Callejero
- Oncology Area, Center for Biomedical Research of La Rioja CIBIR, C/Piqueras 98, Logroño, 26006, Spain.
| | - Alfredo Martínez
- Oncology Area, Center for Biomedical Research of La Rioja CIBIR, C/Piqueras 98, Logroño, 26006, Spain.
| |
Collapse
|
47
|
Javeed N, Sagar G, Dutta SK, Smyrk TC, Lau JS, Bhattacharya S, Truty M, Petersen GM, Kaufman RJ, Chari ST, Mukhopadhyay D. Pancreatic Cancer-Derived Exosomes Cause Paraneoplastic β-cell Dysfunction. Clin Cancer Res 2014. [PMID: 25355928 DOI: 10.1158/1078-0432.ccr-14-2022.pancreatic] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
PURPOSE Pancreatic cancer frequently causes diabetes. We recently proposed adrenomedullin as a candidate mediator of pancreatic β-cell dysfunction in pancreatic cancer. How pancreatic cancer-derived adrenomedullin reaches β cells remote from the cancer to induce β-cell dysfunction is unknown. We tested a novel hypothesis that pancreatic cancer sheds adrenomedullin-containing exosomes into circulation, which are transported to β cells and impair insulin secretion. EXPERIMENTAL METHODS We characterized exosomes from conditioned media of pancreatic cancer cell lines (n = 5) and portal/peripheral venous blood of patients with pancreatic cancer (n = 20). Western blot analysis showed the presence of adrenomedullin in pancreatic cancer-exosomes. We determined the effect of adrenomedullin-containing pancreatic cancer exosomes on insulin secretion from INS-1 β cells and human islets, and demonstrated the mechanism of exosome internalization into β cells. We studied the interaction between β-cell adrenomedullin receptors and adrenomedullin present in pancreatic cancer-exosomes. In addition, the effect of adrenomedullin on endoplasmic reticulum (ER) stress response genes and reactive oxygen/nitrogen species generation in β cells was shown. RESULTS Exosomes were found to be the predominant extracellular vesicles secreted by pancreatic cancer into culture media and patient plasma. Pancreatic cancer-exosomes contained adrenomedullin and CA19-9, readily entered β cells through caveolin-mediated endocytosis or macropinocytosis, and inhibited insulin secretion. Adrenomedullin in pancreatic cancer exosomes interacted with its receptor on β cells. Adrenomedullin receptor blockade abrogated the inhibitory effect of exosomes on insulin secretion. β cells exposed to adrenomedullin or pancreatic cancer exosomes showed upregulation of ER stress genes and increased reactive oxygen/nitrogen species. CONCLUSIONS Pancreatic cancer causes paraneoplastic β-cell dysfunction by shedding adrenomedullin(+)/CA19-9(+) exosomes into circulation that inhibit insulin secretion, likely through adrenomedullin-induced ER stress and failure of the unfolded protein response.
Collapse
Affiliation(s)
- Naureen Javeed
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Gunisha Sagar
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Shamit K Dutta
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Thomas C Smyrk
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Julie S Lau
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Santanu Bhattacharya
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Mark Truty
- Department of Surgery, Mayo Clinic, Rochester, Minnesota
| | - Gloria M Petersen
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | - Randal J Kaufman
- Degenerative Disease Research Program, Sanford Burnham Medical Research Institute, La Jolla, California
| | - Suresh T Chari
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota.
| | | |
Collapse
|
48
|
Lim SY, Ahn SH, Park H, Lee J, Choi K, Choi C, Choi JH, Park EM, Choi YH. Transcriptional regulation of adrenomedullin by oncostatin M in human astroglioma cells: implications for tumor invasion and migration. Sci Rep 2014; 4:6444. [PMID: 25246098 PMCID: PMC4171698 DOI: 10.1038/srep06444] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Accepted: 08/28/2014] [Indexed: 01/05/2023] Open
Abstract
Adrenomedullin (ADM), a secretory peptide with multiple functions in physiological to pathological conditions, is upregulated in several human cancers, including brain, breast, colon, prostate, and lung cancer. However, the molecular mechanisms underlying the regulation of ADM expression in cancerous cells are not fully understood. Here, we report that oncostatin M (OSM), a cytokine belonging to the interleukin-6 family, induces ADM expression in astroglioma cells through induction of signal transducer and activator of transcription-3 (STAT-3) phosphorylation, nuclear translocation, and subsequent DNA binding to the ADM promoter. STAT-3 knockdown decreased OSM-mediated expression of ADM, indicating that ADM expression is regulated by STAT-3 in astroglioma cells. Lastly, scratch wound healing assay showed that astroglioma cell migration was significantly enhanced by ADM peptides. These data suggest that aberrant activation of STAT-3, which is observed in malignant brain tumors, may function as one of the key regulators for ADM expression and glioma invasion.
Collapse
Affiliation(s)
- Seul Ye Lim
- 1] Department of Physiology, Global Top5 Research Program, School of Medicine, Ewha Womans University, Seoul, Korea [2] Tissue Injury Defense Research Center, School of Medicine, Ewha Womans University, Seoul, Korea
| | - So-Hee Ahn
- 1] Department of Physiology, Global Top5 Research Program, School of Medicine, Ewha Womans University, Seoul, Korea [2] Tissue Injury Defense Research Center, School of Medicine, Ewha Womans University, Seoul, Korea
| | - Hyunju Park
- 1] Department of Physiology, Global Top5 Research Program, School of Medicine, Ewha Womans University, Seoul, Korea [2] Tissue Injury Defense Research Center, School of Medicine, Ewha Womans University, Seoul, Korea
| | - Jungsul Lee
- Department of Bio and Brain Engineering, KAIST, Daejeon, Korea
| | - Kyungsun Choi
- Department of Bio and Brain Engineering, KAIST, Daejeon, Korea
| | - Chulhee Choi
- Department of Bio and Brain Engineering, KAIST, Daejeon, Korea
| | - Ji Ha Choi
- 1] Tissue Injury Defense Research Center, School of Medicine, Ewha Womans University, Seoul, Korea [2] Department of Pharmacology, School of Medicine, Ewha Womans University, Seoul, Korea
| | - Eun-Mi Park
- 1] Tissue Injury Defense Research Center, School of Medicine, Ewha Womans University, Seoul, Korea [2] Department of Pharmacology, School of Medicine, Ewha Womans University, Seoul, Korea
| | - Youn-Hee Choi
- 1] Department of Physiology, Global Top5 Research Program, School of Medicine, Ewha Womans University, Seoul, Korea [2] Tissue Injury Defense Research Center, School of Medicine, Ewha Womans University, Seoul, Korea
| |
Collapse
|
49
|
Sudha T, Yalcin M, Lin HY, Elmetwally AM, Nazeer T, Arumugam T, Phillips P, Mousa SA. Suppression of pancreatic cancer by sulfated non-anticoagulant low molecular weight heparin. Cancer Lett 2014; 350:25-33. [PMID: 24769074 DOI: 10.1016/j.canlet.2014.04.016] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 04/14/2014] [Accepted: 04/16/2014] [Indexed: 11/17/2022]
Abstract
Sulfated non-anticoagulant heparins (S-NACHs) might be preferred for potential clinical use in cancer patients without affecting hemostasis as compared to low molecular weight heparins (LMWHs). We investigated anti-tumor effects, anti-angiogenesis effects, and mechanisms of S-NACH in a mouse model of pancreatic cancer as compared to the LMWH tinzaparin. S-NACH or tinzaparin with or without gemcitabine were administered, and tumor luminescent signal intensity, tumor weight, and histopathology were assessed at the termination of the study. S-NACH and LMWH efficiently inhibited tumor growth and metastasis, without any observed bleeding events with S-NACH as compared to tinzaparin. S-NACH distinctly increased tumor necrosis and enhanced gemcitabine response in the mouse pancreatic cancer models. These data suggest the potential implication of S-NACH as a neoadjuvant in pancreatic cancer.
Collapse
Affiliation(s)
- Thangirala Sudha
- The Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY, USA
| | - Murat Yalcin
- The Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY, USA; Department of Physiology, Veterinary Medicine Faculty, Uludag University, Gorukle, Bursa, Turkey
| | - Hung-Yun Lin
- Institute of Cancer Biology and Drug Discovery, Taipei Medical University, Taipei, Taiwan
| | - Ahmed M Elmetwally
- The Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY, USA
| | - Tipu Nazeer
- Department of Pathology, Albany Medical College, Albany, NY, USA
| | - Thiruvengadam Arumugam
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Patricia Phillips
- The Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY, USA
| | - Shaker A Mousa
- The Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY, USA.
| |
Collapse
|
50
|
Ballehaninna UK, Chamberlain RS. Biomarkers for pancreatic cancer: promising new markers and options beyond CA 19-9. Tumour Biol 2013; 34:3279-92. [PMID: 23949878 DOI: 10.1007/s13277-013-1033-3] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Accepted: 07/18/2013] [Indexed: 12/11/2022] Open
Abstract
Pancreatic adenocarcinoma accounts for nearly 90-95% of exocrine malignant tumors of the pancreas. Traditionally, overexpressed proteins/epitopes such as CA 19-9, CA-50, CEA, and many others were being used as pancreatic cancer tumor markers. The main utility of these biomarkers was in the diagnosis of pancreatic cancer as well as to assess response to chemotherapy and to determine prognosis and to predict tumor recurrence. However, these markers had significant limitations such as lack of sensitivity, false-negative results in certain blood groups, as well as false-positive elevation in the presence of obstructive jaundice. To circumvent these limitations, an extraordinary amount of research is being performed to identify an accurate tumor marker or a panel of markers that could aid in the management of the pancreatic cancer. Although this research has identified a large number and different variety of biomarkers, few hold future promise as a preferred marker for pancreatic cancer. This review provides an insight into exciting new areas of pancreatic biomarker research such as salivary, pancreatic juice, and stool markers that can be used as a noninvasive test to identify pancreatic cancer. This manuscript also provides a discussion on newer biomarkers, the role of microRNAs, and pancreatic cancer proteomics, which have the potential to identify a preferred tumor marker for pancreatic adenocarcinoma. This review further elaborates on important genetic changes associated with the development and progression of pancreatic cancer that holds the key for the identification of a sensitive biomarker and which could also serve as a therapeutic target.
Collapse
Affiliation(s)
- Umashankar K Ballehaninna
- Department of Surgery, Saint Barnabas Medical Center, 94, Old Short Hills Road, Livingston, NJ, 07039, USA
| | | |
Collapse
|