1
|
Fan L, Lin Y, Fu Y, Wang J. Small cell lung cancer with liver metastases: from underlying mechanisms to treatment strategies. Cancer Metastasis Rev 2024; 44:5. [PMID: 39585433 DOI: 10.1007/s10555-024-10220-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 11/06/2024] [Indexed: 11/26/2024]
Abstract
Small cell lung cancer (SCLC) represents an aggressive neuroendocrine (NE) tumor within the pulmonary region, characterized by very poor prognoses. Druggable targets for SCLC remain limited, thereby constraining treatment options available to patients. Immuno-chemotherapy has emerged as a pivotal therapeutic strategy for extensive-stage SCLC (ES-SCLC), yet it fails to confer significant efficacy in cases involving liver metastases (LMs) originating from SCLC. Therefore, our attention is directed towards the challenging subset of SCLC patients with LMs. Disease progression of LM-SCLC patients is affected by various factors in the tumor microenvironment (TME), including immune cells, blood vessels, inflammatory mediators, metabolites, and NE substances. Beyond standard immuno-chemotherapy, ongoing efforts to manage LMs in SCLC encompass anti-angiogenic therapy, radiotherapy, microwave ablation (MWA) / radiofrequency ablation (RFA), trans-arterial chemoembolization (TACE), and systemic therapies in conjunction with local interventions. Prospective experimental and clinical investigations into SCLC should prioritize precise and individualized approaches to enhance the prognosis across distinct patient cohorts.
Collapse
Affiliation(s)
- Linjie Fan
- CAMS Key Laboratory of Translational Research on Lung Cancer, State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yiwen Lin
- CAMS Key Laboratory of Translational Research on Lung Cancer, State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yunjie Fu
- School of Basic Medical Sciences, Peking University, Beijing, China
| | - Jie Wang
- CAMS Key Laboratory of Translational Research on Lung Cancer, State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
2
|
Sen C, Koloff CR, Kundu S, Wilkinson DC, Yang JM, Shia DW, Meneses LK, Rickabaugh TM, Gomperts BN. Development of a small cell lung cancer organoid model to study cellular interactions and survival after chemotherapy. Front Pharmacol 2023; 14:1211026. [PMID: 37608896 PMCID: PMC10441219 DOI: 10.3389/fphar.2023.1211026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 06/19/2023] [Indexed: 08/24/2023] Open
Abstract
Introduction: Small-cell-lung-cancer (SCLC) has the worst prognosis of all lung cancers because of a high incidence of relapse after therapy. While lung cancer is the second most common malignancy in the US, only about 10% of cases of lung cancer are SCLC, therefore, it is categorized as a rare and recalcitrant disease. Therapeutic discovery for SCLC has been challenging and the existing pre-clinical models often fail to recapitulate actual tumor pathophysiology. To address this, we developed a bioengineered 3-dimensional (3D) SCLC co-culture organoid model as a phenotypic tool to study SCLC tumor kinetics and SCLC-fibroblast interactions after chemotherapy. Method: We used functionalized alginate microbeads as a scaffold to mimic lung alveolar architecture and co-cultured SCLC cell lines with primary adult lung fibroblasts (ALF). We found that SCLCs in the model proliferated extensively, invaded the microbead scaffold and formed tumors within just 7 days. We compared the bioengineered tumors with patient tumors and found them to recapitulate the pathology and immunophenotyping of the patient tumors. When treated with standard chemotherapy drugs, etoposide and cisplatin, we observed that some of the cells survived the chemotherapy and reformed the tumor in the organoid model. Result and Discussion: Co-culture of the SCLC cells with ALFs revealed that the fibroblasts play a key role in inducing faster and more robust SCLC cell regrowth in the model. This is likely due to a paracrine effect, as conditioned media from the same fibroblasts could also support this accelerated regrowth. This model can be used to study cell-cell interactions and the response to chemotherapy in SCLC and is also scalable and amenable to high throughput phenotypic or targeted drug screening to find new therapeutics for SCLC.
Collapse
Affiliation(s)
- Chandani Sen
- Department of Pediatrics, David Geffen School of Medicine, UCLA Children’s Discovery and Innovation Institute, Mattel Children’s Hospital, University of California, Los Angeles, CA, United States
| | - Caroline R. Koloff
- Department of Pediatrics, David Geffen School of Medicine, UCLA Children’s Discovery and Innovation Institute, Mattel Children’s Hospital, University of California, Los Angeles, CA, United States
| | | | - Dan C. Wilkinson
- Department of Pediatrics, David Geffen School of Medicine, UCLA Children’s Discovery and Innovation Institute, Mattel Children’s Hospital, University of California, Los Angeles, CA, United States
| | - Juliette M. Yang
- Department of Pediatrics, David Geffen School of Medicine, UCLA Children’s Discovery and Innovation Institute, Mattel Children’s Hospital, University of California, Los Angeles, CA, United States
| | - David W. Shia
- Department of Pediatrics, David Geffen School of Medicine, UCLA Children’s Discovery and Innovation Institute, Mattel Children’s Hospital, University of California, Los Angeles, CA, United States
| | - Luisa K. Meneses
- Department of Pediatrics, David Geffen School of Medicine, UCLA Children’s Discovery and Innovation Institute, Mattel Children’s Hospital, University of California, Los Angeles, CA, United States
| | - Tammy M. Rickabaugh
- Department of Pediatrics, David Geffen School of Medicine, UCLA Children’s Discovery and Innovation Institute, Mattel Children’s Hospital, University of California, Los Angeles, CA, United States
| | - Brigitte N. Gomperts
- Department of Pediatrics, David Geffen School of Medicine, UCLA Children’s Discovery and Innovation Institute, Mattel Children’s Hospital, University of California, Los Angeles, CA, United States
- Pulmonary Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, United States
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, United States
- Eli and Edythe Broad Stem Cell Research Center, University of California, Los Angeles, CA, United States
| |
Collapse
|
3
|
Groves SM, Panchy N, Tyson DR, Harris LA, Quaranta V, Hong T. Involvement of Epithelial-Mesenchymal Transition Genes in Small Cell Lung Cancer Phenotypic Plasticity. Cancers (Basel) 2023; 15:1477. [PMID: 36900269 PMCID: PMC10001072 DOI: 10.3390/cancers15051477] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 02/16/2023] [Accepted: 02/20/2023] [Indexed: 03/03/2023] Open
Abstract
Small cell lung cancer (SCLC) is an aggressive cancer recalcitrant to treatment, arising predominantly from epithelial pulmonary neuroendocrine (NE) cells. Intratumor heterogeneity plays critical roles in SCLC disease progression, metastasis, and treatment resistance. At least five transcriptional SCLC NE and non-NE cell subtypes were recently defined by gene expression signatures. Transition from NE to non-NE cell states and cooperation between subtypes within a tumor likely contribute to SCLC progression by mechanisms of adaptation to perturbations. Therefore, gene regulatory programs distinguishing SCLC subtypes or promoting transitions are of great interest. Here, we systematically analyze the relationship between SCLC NE/non-NE transition and epithelial to mesenchymal transition (EMT)-a well-studied cellular process contributing to cancer invasiveness and resistance-using multiple transcriptome datasets from SCLC mouse tumor models, human cancer cell lines, and tumor samples. The NE SCLC-A2 subtype maps to the epithelial state. In contrast, SCLC-A and SCLC-N (NE) map to a partial mesenchymal state (M1) that is distinct from the non-NE, partial mesenchymal state (M2). The correspondence between SCLC subtypes and the EMT program paves the way for further work to understand gene regulatory mechanisms of SCLC tumor plasticity with applicability to other cancer types.
Collapse
Affiliation(s)
- Sarah M. Groves
- Department of Biochemistry, Vanderbilt University, Nashville, TN 37235, USA
| | - Nicholas Panchy
- Department of Biochemistry & Cellular and Molecular Biology, The University of Tennessee, Knoxville, TN 37996, USA
| | - Darren R. Tyson
- Department of Biochemistry, Vanderbilt University, Nashville, TN 37235, USA
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37235, USA
| | - Leonard A. Harris
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR 72701, USA
- Interdisciplinary Graduate Program in Cell and Molecular Biology, University of Arkansas, Fayetteville, AR 72701, USA
- Cancer Biology Program, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Vito Quaranta
- Department of Biochemistry, Vanderbilt University, Nashville, TN 37235, USA
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37235, USA
| | - Tian Hong
- Department of Biochemistry & Cellular and Molecular Biology, The University of Tennessee, Knoxville, TN 37996, USA
- National Institute for Mathematical and Biological Synthesis, Knoxville, TN 37996, USA
| |
Collapse
|
4
|
Sen C, Koloff C, Kundu S, Wilkinson DC, Yang J, Shia DW, Meneses LK, Rickabaugh TM, Gomperts BN. Small cell lung cancer co-culture organoids provide insights into cancer cell survival after chemotherapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.03.522668. [PMID: 36711908 PMCID: PMC9881945 DOI: 10.1101/2023.01.03.522668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Small-cell-lung-cancer (SCLC) has the worst prognosis of all lung cancers because of a high incidence of relapse after therapy. We developed a bioengineered 3-dimensional (3D) SCLC co-culture organoid as a phenotypic tool to study SCLC tumor kinetics and SCLC-fibroblast interactions during relapse. We used functionalized alginate microbeads as a scaffold to mimic lung alveolar architecture and co-cultured SCLC cell lines with primary adult lung fibroblasts (ALF). We found that SCLCs in the model proliferated extensively, invaded the microbead scaffold and formed tumors within just 7 days. We compared the bioengineered tumors with patient tumors and found them to recapitulate the pathology and immunophenotyping of the patient tumors better than the PDX model developed from the same SCLC cell line. When treated with standard chemotherapy drugs, etoposide and cisplatin, the organoid recapitulated relapse after chemotherapy. Co-culture of the SCLC cells with ALFs revealed that the fibroblasts play a key role in inducing faster and more robust SCLC cell regrowth in the model. This was a paracrine effect as conditioned medium from the same fibroblasts was responsible for this accelerated cell regrowth. This model is also amenable to high throughput phenotypic or targeted drug screening to find new therapeutics for SCLC.
Collapse
|
5
|
van der Valk ES, Kleinendorst L, Delhanty PJD, van der Voorn B, Visser JA, van Haelst MM, de Graaff LCG, Huisman M, White A, Ito S, Wakamatsu K, de Rijke YB, van den Akker ELT, Iyer AM, van Rossum EFC. Obesity and Hyperphagia With Increased Defective ACTH: A Novel POMC Variant. J Clin Endocrinol Metab 2022; 107:e3699-e3704. [PMID: 35737586 PMCID: PMC9797039 DOI: 10.1210/clinem/dgac342] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Indexed: 01/05/2023]
Abstract
OBJECTIVE Patients with pro-opiomelanocortin (POMC) defects generally present with early-onset obesity, hyperphagia, hypopigmentation and adrenocorticotropin (ACTH) deficiency. Rodent models suggest that adequate cleavage of ACTH to α-melanocortin-stimulating hormone (α-MSH) and desacetyl-α-melanocortin-stimulating hormone (d-α-MSH) by prohormone convertase 2 at the KKRR region is required for regulating food intake and energy balance. METHODS We present 2 sisters with a novel POMC gene variant, leading to an ACTH defect at the prohormone convertase 2 cleavage site, and performed functional studies of this variant. RESULTS The patients had obesity, hyperphagia and hypocortisolism, with markerly raised levels of ACTH but unaffected pigmentation. Their ACTH has reduced potency to stimulate the melanocortin (MC) 2 receptor, explaining their hypocortisolism. CONCLUSION The hyperphagia and obesity support evidence that adequate cleavage of ACTH to α-MSH and d-α-MSH is also required in humans for feeding control.
Collapse
Affiliation(s)
- Eline S van der Valk
- Obesity Centre CGG, Erasmus University Medical Center Rotterdam, 3000 CA Rotterdam, the Netherlands
- Department of Internal Medicine, Division of Endocrinology, Erasmus University Medical Center Rotterdam, 3000 CA Rotterdam, the Netherlands
| | | | | | - Bibian van der Voorn
- Obesity Centre CGG, Erasmus University Medical Center Rotterdam, 3000 CA Rotterdam, the Netherlands
- Department of Internal Medicine, Division of Endocrinology, Erasmus University Medical Center Rotterdam, 3000 CA Rotterdam, the Netherlands
- Department of Pediatrics, Erasmus University Medical Center, 3000 CA Rotterdam, the Netherlands
| | - Jenny A Visser
- Obesity Centre CGG, Erasmus University Medical Center Rotterdam, 3000 CA Rotterdam, the Netherlands
- Department of Internal Medicine, Division of Endocrinology, Erasmus University Medical Center Rotterdam, 3000 CA Rotterdam, the Netherlands
| | - M M van Haelst
- Department of Clinical Genetics, Amsterdam UMC, University of Amsterdam, 1100 DD Amsterdam, the Netherlands
| | - Laura C G de Graaff
- Department of Internal Medicine, Division of Endocrinology, Erasmus University Medical Center Rotterdam, 3000 CA Rotterdam, the Netherlands
| | - Martin Huisman
- Department of Internal Medicine, Division of Endocrinology, Erasmus University Medical Center Rotterdam, 3000 CA Rotterdam, the Netherlands
| | - Anne White
- Divison of Diabetes, Endocrinology & Gastroenterology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK
| | - Shosuke Ito
- Institute for Melanin Chemistry, Fujita Health University, Toyoake, 470-1192, Japan
| | - Kazumasa Wakamatsu
- Institute for Melanin Chemistry, Fujita Health University, Toyoake, 470-1192, Japan
| | - Yolanda B de Rijke
- Department of Clinical Chemistry, Erasmus University Medical Center Rotterdam, 3000 CA Rotterdam, the Netherlands
| | - Erica L T van den Akker
- Obesity Centre CGG, Erasmus University Medical Center Rotterdam, 3000 CA Rotterdam, the Netherlands
- Department of Pediatrics, Erasmus University Medical Center, 3000 CA Rotterdam, the Netherlands
| | - Anand M Iyer
- Obesity Centre CGG, Erasmus University Medical Center Rotterdam, 3000 CA Rotterdam, the Netherlands
- Department of Internal Medicine, Division of Endocrinology, Erasmus University Medical Center Rotterdam, 3000 CA Rotterdam, the Netherlands
| | - Elisabeth F C van Rossum
- Correspondence: Elisabeth F. C. van Rossum, MD, PhD, Obesity Center CGG (Centrum Gezond Gewicht), Erasmus MC, University Medical Center Rotterdam, the Netherlands, Rm Rg-5. P. O. Box 2400, 3000 CA Rotterdam, the Netherlands.
| |
Collapse
|
6
|
Liu Y, Xia J, McKay J, Tsavachidis S, Xiao X, Spitz MR, Cheng C, Byun J, Hong W, Li Y, Zhu D, Song Z, Rosenberg SM, Scheurer ME, Kheradmand F, Pikielny CW, Lusk CM, Schwartz AG, Wistuba II, Cho MH, Silverman EK, Bailey-Wilson J, Pinney SM, Anderson M, Kupert E, Gaba C, Mandal D, You M, de Andrade M, Yang P, Liloglou T, Davies MPA, Lissowska J, Swiatkowska B, Zaridze D, Mukeria A, Janout V, Holcatova I, Mates D, Stojsic J, Scelo G, Brennan P, Liu G, Field JK, Hung RJ, Christiani DC, Amos CI. Rare deleterious germline variants and risk of lung cancer. NPJ Precis Oncol 2021; 5:12. [PMID: 33594163 PMCID: PMC7887261 DOI: 10.1038/s41698-021-00146-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 12/11/2020] [Indexed: 01/19/2023] Open
Abstract
Recent studies suggest that rare variants exhibit stronger effect sizes and might play a crucial role in the etiology of lung cancers (LC). Whole exome plus targeted sequencing of germline DNA was performed on 1045 LC cases and 885 controls in the discovery set. To unveil the inherited causal variants, we focused on rare and predicted deleterious variants and small indels enriched in cases or controls. Promising candidates were further validated in a series of 26,803 LCs and 555,107 controls. During discovery, we identified 25 rare deleterious variants associated with LC susceptibility, including 13 reported in ClinVar. Of the five validated candidates, we discovered two pathogenic variants in known LC susceptibility loci, ATM p.V2716A (Odds Ratio [OR] 19.55, 95%CI 5.04-75.6) and MPZL2 p.I24M frameshift deletion (OR 3.88, 95%CI 1.71-8.8); and three in novel LC susceptibility genes, POMC c.*28delT at 3' UTR (OR 4.33, 95%CI 2.03-9.24), STAU2 p.N364M frameshift deletion (OR 4.48, 95%CI 1.73-11.55), and MLNR p.Q334V frameshift deletion (OR 2.69, 95%CI 1.33-5.43). The potential cancer-promoting role of selected candidate genes and variants was further supported by endogenous DNA damage assays. Our analyses led to the identification of new rare deleterious variants with LC susceptibility. However, in-depth mechanistic studies are still needed to evaluate the pathogenic effects of these specific alleles.
Collapse
Grants
- R01 CA060691 NCI NIH HHS
- U19 CA203654 NCI NIH HHS
- R01 CA084354 NCI NIH HHS
- R01 HL110883 NHLBI NIH HHS
- U01 CA076293 NCI NIH HHS
- R01 CA080127 NCI NIH HHS
- R01 CA141769 NCI NIH HHS
- P30 ES006096 NIEHS NIH HHS
- P50 CA090578 NCI NIH HHS
- P30 CA022453 NCI NIH HHS
- S10 RR024574 NCRR NIH HHS
- HHSN261201300011C NCI NIH HHS
- R01 CA134682 NCI NIH HHS
- R01 CA134433 NCI NIH HHS
- R01 HL113264 NHLBI NIH HHS
- R01 HL082487 NHLBI NIH HHS
- R01 CA250905 NCI NIH HHS
- U19 CA148127 NCI NIH HHS
- P20 GM103534 NIGMS NIH HHS
- R01 CA092824 NCI NIH HHS
- R01 CA087895 NCI NIH HHS
- U01 HL089897 NHLBI NIH HHS
- K07 CA181480 NCI NIH HHS
- HHSN268201100011I NHLBI NIH HHS
- HHSN268201100011C NHLBI NIH HHS
- R01 CA127219 NCI NIH HHS
- R01 CA074386 NCI NIH HHS
- P30 CA023108 NCI NIH HHS
- U01 HL089856 NHLBI NIH HHS
- P30 ES030285 NIEHS NIH HHS
- P30 CA125123 NCI NIH HHS
- DP1 AG072751 NIA NIH HHS
- U01 CA243483 NCI NIH HHS
- HHSN268200782096C NHLBI NIH HHS
- HHSN268201200007C NHLBI NIH HHS
- N01HG65404 NHGRI NIH HHS
- R35 GM122598 NIGMS NIH HHS
- U01 CA209414 NCI NIH HHS
- R03 CA077118 NCI NIH HHS
- 001 World Health Organization
- DP1 CA174424 NCI NIH HHS
- This work was supported by grants from the National Institutes of Health (R01CA127219, R01CA141769, R01CA060691, R01CA87895, R01CA80127, R01CA84354, R01CA134682, R01CA134433, R01CA074386, R01CA092824, R01CA250905, R01HL113264, R01HL082487, R01HL110883, R03CA77118, P20GM103534, P30CA125123, P30CA023108, P30CA022453, P30ES006096, P50CA090578, U01CA243483, U01HL089856, U01HL089897, U01CA76293, U19CA148127, U01CA209414, K07CA181480, N01-HG-65404, HHSN268200782096C, HHSN261201300011I, HHSN268201100011, HHSN268201 200007C, DP1-CA174424, DP1-AG072751, CA125123, RR024574, Intramural Research Program of the National Human Genome Research Institute (JEB-W), and Herrick Foundation. Dr. Amos is an Established Research Scholar of the Cancer Prevention Research Institute of Texas (RR170048). We also want to acknowledge the Cytometry and Cell Sorting Core support by the Cancer Prevention and Research Institute of Texas Core Facility (RP180672). At Toronto, the study is supported by The Canadian Cancer Society Research Institute (# 020214) to R. H., Ontario Institute for Cancer Research to R. H, and the Alan Brown Chair to G. L. and Lusi Wong Programs at the Princess Margaret Hospital Foundation. The Liverpool Lung Project is supported by Roy Castle Lung Cancer Foundation.
Collapse
Affiliation(s)
- Yanhong Liu
- Dan L. Duncan Comprehensive Cancer Center, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Jun Xia
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, USA
| | - James McKay
- International Agency for Research on Cancer, Lyon, France
| | - Spiridon Tsavachidis
- Dan L. Duncan Comprehensive Cancer Center, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Xiangjun Xiao
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, USA
| | - Margaret R Spitz
- Dan L. Duncan Comprehensive Cancer Center, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Chao Cheng
- Dan L. Duncan Comprehensive Cancer Center, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, USA
| | - Jinyoung Byun
- Dan L. Duncan Comprehensive Cancer Center, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, USA
| | - Wei Hong
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, USA
| | - Yafang Li
- Dan L. Duncan Comprehensive Cancer Center, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, USA
| | - Dakai Zhu
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, USA
| | - Zhuoyi Song
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, USA
| | - Susan M Rosenberg
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Michael E Scheurer
- Dan L. Duncan Comprehensive Cancer Center, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Farrah Kheradmand
- Dan L. Duncan Comprehensive Cancer Center, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
- Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, USA
| | - Claudio W Pikielny
- Department of Biomedical Data Science, Geisel School of Medicine, Dartmouth College, Lebanon, NH, USA
| | - Christine M Lusk
- Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Ann G Schwartz
- Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Ignacio I Wistuba
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael H Cho
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Edwin K Silverman
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | | | - Susan M Pinney
- University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | | | - Elena Kupert
- University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Colette Gaba
- The University of Toledo College of Medicine, Toledo, OH, USA
| | - Diptasri Mandal
- Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Ming You
- Medical College of Wisconsin, Milwaukee, WI, USA
| | | | - Ping Yang
- Mayo Clinic College of Medicine, Scottsdale, AZ, USA
| | - Triantafillos Liloglou
- Roy Castle Lung Cancer Research Programme, The University of Liverpool, Department of Molecular and Clinical Cancer Medicine, Liverpool, UK
| | - Michael P A Davies
- Roy Castle Lung Cancer Research Programme, The University of Liverpool, Department of Molecular and Clinical Cancer Medicine, Liverpool, UK
| | - Jolanta Lissowska
- M. Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Beata Swiatkowska
- Nofer Institute of Occupational Medicine, Department of Environmental Epidemiology, Lodz, Poland
| | - David Zaridze
- Russian N.N. Blokhin Cancer Research Centre, Moscow, Russian Federation
| | - Anush Mukeria
- Russian N.N. Blokhin Cancer Research Centre, Moscow, Russian Federation
| | - Vladimir Janout
- Faculty of Health Sciences, Palacky University, Olomouc, Czech Republic
| | - Ivana Holcatova
- Institute of Public Health and Preventive Medicine, Charles University, 2nd Faculty of Medicine, Prague, Czech Republic
| | - Dana Mates
- National Institute of Public Health, Bucharest, Romania
| | - Jelena Stojsic
- Department of Thoracopulmonary Pathology, Service of Pathology, Clinical Center of Serbia, Belgrade, Serbia
| | | | - Paul Brennan
- International Agency for Research on Cancer, Lyon, France
| | - Geoffrey Liu
- Princess Margaret Cancer Center, Toronto, ON, Canada
| | - John K Field
- Roy Castle Lung Cancer Research Programme, The University of Liverpool, Department of Molecular and Clinical Cancer Medicine, Liverpool, UK
| | - Rayjean J Hung
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | | | - Christopher I Amos
- Dan L. Duncan Comprehensive Cancer Center, Department of Medicine, Baylor College of Medicine, Houston, TX, USA.
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
7
|
Jarvela TS, Surbhi , Shakya M, Bachor T, White A, Low MJ, Lindberg I. Reduced Stability and pH-Dependent Activity of a Common Obesity-Linked PCSK1 Polymorphism, N221D. Endocrinology 2019; 160:2630-2645. [PMID: 31504391 PMCID: PMC6892424 DOI: 10.1210/en.2019-00418] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 08/26/2019] [Indexed: 02/06/2023]
Abstract
Common mutations in the human prohormone convertase (PC)1/3 gene (PCKSI) are linked to increased risk of obesity. Previous work has shown that the rs6232 single-nucleotide polymorphism (N221D) results in slightly decreased activity, although whether this decrease underlies obesity risk is not clear. We observed significantly decreased activity of the N221D PC1/3 enzyme at the pH of the trans-Golgi network; at this pH, the mutant enzyme was less stable than wild-type enzyme. Recombinant N221D PC1/3 also showed enhanced susceptibility to heat stress. Enhanced susceptibility to tunicamycin-induced endoplasmic reticulum stress was observed in AtT-20/PC2 cell clones in which murine PC1/3 was replaced by human N221D PC1/3, as compared with wild-type human PC1/3. However, N221D PC1/3-expressing AtT-20/PC2 clones processed proopiomelanocortin to α-MSH similarly to wild-type PC1/3. We also generated a CRISPR-edited mouse line expressing the N221D mutation in the PCKSI gene. When homozygous N221D mice were fed either a standard or a high-fat diet, we found no increase in body weight compared with their wild-type sibling controls. Sexual dimorphism was observed in pituitary ACTH for both genotypes, with females exhibiting lower levels of pituitary ACTH. In contrast, hypothalamic α-MSH content for both genotypes was higher in females compared with males. Hypothalamic corticotropin-like intermediate peptide content was higher in wild-type females compared with wild-type, but not N221D, males. Taken together, these data suggest that the increased obesity risk linked to the N221D allele in humans may be due in part to PC1/3-induced loss of resilience to stressors rather than strictly to decreased enzymatic activity on peptide precursors.
Collapse
Affiliation(s)
- Timothy S Jarvela
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Surbhi
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Manita Shakya
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Tomas Bachor
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Anne White
- Division of Diabetes, Endocrinology and Gastroenterology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Malcolm J Low
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Iris Lindberg
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland
- Correspondence: Iris Lindberg, PhD, Department of Anatomy and Neurobiology, University of Maryland School of Medicine, 20 Penn Street, Room S267, Baltimore, Maryland 21210. E-mail:
| |
Collapse
|
8
|
Kirwan P, Kay RG, Brouwers B, Herranz-Pérez V, Jura M, Larraufie P, Jerber J, Pembroke J, Bartels T, White A, Gribble FM, Reimann F, Farooqi IS, O'Rahilly S, Merkle FT. Quantitative mass spectrometry for human melanocortin peptides in vitro and in vivo suggests prominent roles for β-MSH and desacetyl α-MSH in energy homeostasis. Mol Metab 2018; 17:82-97. [PMID: 30201275 PMCID: PMC6197775 DOI: 10.1016/j.molmet.2018.08.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 08/08/2018] [Accepted: 08/16/2018] [Indexed: 12/23/2022] Open
Abstract
OBJECTIVE The lack of pro-opiomelanocortin (POMC)-derived melanocortin peptides results in hypoadrenalism and severe obesity in both humans and rodents that is treatable with synthetic melanocortins. However, there are significant differences in POMC processing between humans and rodents, and little is known about the relative physiological importance of POMC products in the human brain. The aim of this study was to determine which POMC-derived peptides are present in the human brain, to establish their relative concentrations, and to test if their production is dynamically regulated. METHODS We analysed both fresh post-mortem human hypothalamic tissue and hypothalamic neurons derived from human pluripotent stem cells (hPSCs) using liquid chromatography tandem mass spectrometry (LC-MS/MS) to determine the sequence and quantify the production of hypothalamic neuropeptides, including those derived from POMC. RESULTS In both in vitro and in vivo hypothalamic cells, LC-MS/MS revealed the sequence of hundreds of neuropeptides as a resource for the field. Although the existence of β-melanocyte stimulating hormone (MSH) is controversial, we found that both this peptide and desacetyl α-MSH (d-α-MSH) were produced in considerable excess of acetylated α-MSH. In hPSC-derived hypothalamic neurons, these POMC derivatives were appropriately trafficked, secreted, and their production was significantly (P < 0.0001) increased in response to the hormone leptin. CONCLUSIONS Our findings challenge the assumed pre-eminence of α-MSH and suggest that in humans, d-α-MSH and β-MSH are likely to be the predominant physiological products acting on melanocortin receptors.
Collapse
Affiliation(s)
- Peter Kirwan
- Metabolic Research Laboratories and Medical Research Council Metabolic Diseases Unit, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK; The Anne McLaren Laboratory for Regenerative Medicine, Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, CB2 0SZ, UK
| | - Richard G Kay
- Metabolic Research Laboratories and Medical Research Council Metabolic Diseases Unit, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Bas Brouwers
- Metabolic Research Laboratories and Medical Research Council Metabolic Diseases Unit, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK; The Anne McLaren Laboratory for Regenerative Medicine, Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, CB2 0SZ, UK
| | - Vicente Herranz-Pérez
- Laboratory of Comparative Neurobiology, Cavanilles Institute of Biodiversity and Evolutionary Biology, University of Valencia, CIBERNED, 46980 Valencia, Spain; Predepartamental Unit of Medicine, Faculty of Health Sciences, Universitat Jaume I, 12071 Castelló de la Plana, Spain
| | - Magdalena Jura
- Metabolic Research Laboratories and Medical Research Council Metabolic Diseases Unit, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK; The Anne McLaren Laboratory for Regenerative Medicine, Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, CB2 0SZ, UK
| | - Pierre Larraufie
- Metabolic Research Laboratories and Medical Research Council Metabolic Diseases Unit, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Julie Jerber
- The Anne McLaren Laboratory for Regenerative Medicine, Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, CB2 0SZ, UK; Open Targets, Wellcome Trust Sanger Institute, Hinxton, CB10 1SA, UK
| | - Jason Pembroke
- LGC Ltd., Newmarket Road, Fordham, Cambridgeshire, CB7 5WW, UK
| | - Theresa Bartels
- The Anne McLaren Laboratory for Regenerative Medicine, Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, CB2 0SZ, UK
| | - Anne White
- Division of Diabetes, Endocrinology and Gastroenterology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Fiona M Gribble
- Metabolic Research Laboratories and Medical Research Council Metabolic Diseases Unit, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Frank Reimann
- Metabolic Research Laboratories and Medical Research Council Metabolic Diseases Unit, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - I Sadaf Farooqi
- Metabolic Research Laboratories and Medical Research Council Metabolic Diseases Unit, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Stephen O'Rahilly
- Metabolic Research Laboratories and Medical Research Council Metabolic Diseases Unit, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Florian T Merkle
- Metabolic Research Laboratories and Medical Research Council Metabolic Diseases Unit, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK; The Anne McLaren Laboratory for Regenerative Medicine, Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, CB2 0SZ, UK.
| |
Collapse
|
9
|
Zhu R, Yang X, Xue X, Shen M, Chen F, Chen X, Tsai Y, Keng PC, Chen Y, Lee SO, Chen Y. RETRACTED: Neuroendocrine differentiation contributes to radioresistance development and metastatic potential increase in non-small cell lung cancer. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2018; 1865:1878-1890. [PMID: 30262435 DOI: 10.1016/j.bbamcr.2018.09.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 09/11/2018] [Accepted: 09/13/2018] [Indexed: 12/29/2022]
Abstract
This article has been retracted: please see Elsevier Policy on Article Withdrawal (http://www.elsevier.com/locate/withdrawalpolicy).
This article has been retracted at the request of the authors and their institute. The BBA Editor-in-Chief has agreed to retract the paper.
In this paper, there were two errors identified to the journal by the authors: The first error was in Western blot gel band images of Fig. 4A (p-MAPK, MAPK, p-Erk, and Stat3) and the 8 gel band images of Fig. 4G. The second error was in the cell culture images of Figures 3F, 3J, and 4E.
The authors state that these errors were due to uploading mistakes in the preparation of the manuscript. The authors apologize for these errors and any inconvenience caused.
The Editor-in-Chief initially agreed to retract the paper based on the identification of these two errors. Readers are able to see further discussion of the paper on the PubPeer site here: https://pubpeer.com/publications/569EB2CE7A7335D7F3F8F3FF310936
Collapse
Affiliation(s)
- Rongying Zhu
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA; Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, PR China
| | - Xiaodong Yang
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, PR China
| | - Xiang Xue
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, PR China
| | - Mingjing Shen
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, PR China
| | - Feng Chen
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Xiaodong Chen
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Ying Tsai
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Peter C Keng
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Yongbing Chen
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, PR China
| | - Soo Ok Lee
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA.
| | - Yuhchyau Chen
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA.
| |
Collapse
|
10
|
Lim SL, Jia Z, Lu Y, Zhang H, Ng CT, Bay BH, Shen HM, Ong CN. Metabolic signatures of four major histological types of lung cancer cells. Metabolomics 2018; 14:118. [PMID: 30830374 DOI: 10.1007/s11306-018-1417-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 08/21/2018] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Histologically lung cancer is classified into four major types: adenocarcinoma (Ad), squamous cell carcinoma (SqCC), large cell carcinoma (LCC), and small cell lung cancer (SCLC). Presently, our understanding of cellular metabolism among them is still not clear. OBJECTIVES The goal of this study was to assess the cellular metabolic profiles across these four types of lung cancer using an untargeted metabolomics approach. METHODS Six lung cancer cell lines, viz., Ad (A549 and HCC827), SqCC (NCl-H226 and NCl-H520), LCC (NCl-H460), and SCLC (NCl-H526), were analyzed using liquid chromatography quadrupole time-of-flight mass spectrometry, with normal human small airway epithelial cells (SAEC) as the control group. The principal component analysis (PCA) was performed to identify the metabolic signatures that had characteristic alterations in each histological type. Further, a metabolite set enrichment analysis was performed for pathway analysis. RESULTS Compared to the SAEC, 31, 27, 34, 34, 32, and 39 differential metabolites mainly in relation to nucleotides, amino acid, and fatty acid metabolism were identified in A549, HCC827, NCl-H226, NCl-H520, NCl-H460, and NCl-H526 cells, respectively. The metabolic signatures allowed the six cancerous cell lines to be clearly separated in a PCA score plot. CONCLUSION The metabolic signatures are unique to each histological type, and appeared to be related to their cell-of-origin and mutation status. The changes are useful for assessing the metabolic characteristics of lung cancer, and offer potential for the establishment of novel diagnostic tools for different origin and oncogenic mutation of lung cancer.
Collapse
Affiliation(s)
- Swee Ling Lim
- Saw Swee Hock School of Public Health, National University of Singapore, 12 Science Drive 2, #11-01, Tahir Foundation Building, Singapore, 117549, Singapore
| | - Zhunan Jia
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore, 117456, Singapore
- NUS Nanoscience & Nanotechnology Initiative, National University of Singapore, Singapore, 117411, Singapore
| | - Yonghai Lu
- Saw Swee Hock School of Public Health, National University of Singapore, 12 Science Drive 2, #11-01, Tahir Foundation Building, Singapore, 117549, Singapore.
| | - Hui Zhang
- NUS Environmental Research Institute, National University of Singapore, #02-01, T-Lab Building, 5A Engineering Drive 1, Singapore, 117411, Singapore
| | - Cheng Teng Ng
- NUS Environmental Research Institute, National University of Singapore, #02-01, T-Lab Building, 5A Engineering Drive 1, Singapore, 117411, Singapore
| | - Boon Huat Bay
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Han Ming Shen
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117593, Singapore
| | - Choon Nam Ong
- Saw Swee Hock School of Public Health, National University of Singapore, 12 Science Drive 2, #11-01, Tahir Foundation Building, Singapore, 117549, Singapore.
- NUS Environmental Research Institute, National University of Singapore, #02-01, T-Lab Building, 5A Engineering Drive 1, Singapore, 117411, Singapore.
| |
Collapse
|
11
|
Mendieta I, Nuñez-Anita RE, Pérez-Sánchez G, Pavón L, Rodríguez-Cruz A, García-Alcocer G, Berumen LC. Effect of A549 neuroendocrine differentiation on cytotoxic immune response. Endocr Connect 2018; 7:791-802. [PMID: 29700099 PMCID: PMC5987362 DOI: 10.1530/ec-18-0145] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 04/26/2018] [Indexed: 12/29/2022]
Abstract
The present study was designed to determine the effects of factors secreted by the lung adenocarcinoma cell line with the neuroendocrine phenotype, A549NED, on cytotoxic T lymphocytes (CTLs) activity in vitro A perspective that integrates the nervous, endocrine and immune system in cancer research is essential to understand the complexity of dynamic interactions in tumours. Extensive clinical research suggests that neuroendocrine differentiation (NED) is correlated with worse patient outcomes; however, little is known regarding the effects of neuroendocrine factors on the communication between the immune system and neoplastic cells. The human lung cancer cell line A549 was induced to NED (A549NED) using cAMP-elevating agents. The A549NED cells showed changes in cell morphology, an inhibition of proliferation, an overexpression of chromogranin and a differential pattern of biogenic amine production (decreased dopamine and increased serotonin [5-HT] levels). Using co-cultures to determine the cytolytic CTLs activity on target cells, we showed that the acquisition of NED inhibits the decrease in the viability of the target cells and release of fluorescence. Additionally, the conditioned medium of A549NED and 5-HT considerably decreased the viability and proliferation of the Jurkat cells after 24 h. Thus, our study successfully generated a neuroendocrine phenotype from the A549 cell line. In co-cultures with CTLs, the pattern of secretion by A549NED impaired the proliferation and cytotoxic activity of CTLs, which might be partly explained by the increased release of 5-HT.
Collapse
Affiliation(s)
- Irasema Mendieta
- Facultad de QuímicaUniversidad Autónoma de Querétaro, Querétaro, Mexico
| | - Rosa Elvira Nuñez-Anita
- Facultad de Medicina Veterinaria y ZootecniaUniversidad Michoacana de San Nicolás Hidalgo, Morelia, Michoacán, Mexico
| | - Gilberto Pérez-Sánchez
- Departmento de PsicoimunologíaInstituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Ciudad de México, Mexico
| | - Lenin Pavón
- Departmento de PsicoimunologíaInstituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Ciudad de México, Mexico
| | | | | | | |
Collapse
|
12
|
Šlekienė L, Stakišaitis D, Balnytė I, Valančiūtė A. Sodium Valproate Inhibits Small Cell Lung Cancer Tumor Growth on the Chicken Embryo Chorioallantoic Membrane and Reduces the p53 and EZH2 Expression. Dose Response 2018; 16:1559325818772486. [PMID: 29760602 PMCID: PMC5944146 DOI: 10.1177/1559325818772486] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 03/26/2018] [Accepted: 03/27/2018] [Indexed: 12/30/2022] Open
Abstract
The study aims to test the effect of different sodium valproate (NaVP) doses on small cell lung cancer NCI-H146 cells tumor in chicken embryo chorioallantoic membrane (CAM) model. Xenografts were investigated in the following groups: nontreated control and 5 groups treated with different NaVP doses (2, 3, 4, 6, and 8 mmol/L). Invasion of tumors into CAM in the nontreated group reached 76%. Tumors treated with 8 mmol/L NaVP doses significantly differed in tumor invasion frequency from the control and those treated with 2 mmol/L (P < .01). The calculated probability of 50% tumor noninvasion into CAM was when tumors were treated with 4 mmol/L of NaVP. Number of p53-positive cells in tumors was significantly reduced when treated with NaVP doses from 3 to 8 mmol/L as compared with control; number of EZH2-positive cells in control significantly differed from all NaVP-treated groups. No differences in p53- and EZH2-positive cell numbers were found among 4, 6, and 8 mmol/L NaVP-treated groups. Invaded tumors had an increased N-cadherin and reduced E-cadherin expression. The results indicate the increasing NaVP dose to be able to inhibit tumors progression. Expression of p53 and EZH2 may be promising target markers of therapeutic efficacy evaluation.
Collapse
Affiliation(s)
- Lina Šlekienė
- Department of Histology and Embryology, Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Donatas Stakišaitis
- Department of Histology and Embryology, Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania.,Laboratory of Molecular Oncology, National Cancer Institute, Vilnius, Lithuania
| | - Ingrida Balnytė
- Department of Histology and Embryology, Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Angelija Valančiūtė
- Department of Histology and Embryology, Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania
| |
Collapse
|
13
|
Bryant JL, Gieling RG, Meredith SL, Allen TJ, Walker L, Telfer BA, Supuran CT, Williams KJ, White A. Novel carbonic anhydrase IX-targeted therapy enhances the anti-tumour effects of cisplatin in small cell lung cancer. Int J Cancer 2017; 142:191-201. [DOI: 10.1002/ijc.31042] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Accepted: 08/28/2017] [Indexed: 12/12/2022]
Affiliation(s)
- Jennifer L Bryant
- Division of Diabetes, Endocrinology & Gastroenterology; University of Manchester; Manchester United Kingdom
- Division of Pharmacy & Optometry, School of Biology, Medicine and Health; University of Manchester; Manchester United Kingdom
| | - Roben G Gieling
- Division of Pharmacy & Optometry, School of Biology, Medicine and Health; University of Manchester; Manchester United Kingdom
| | - Suzanne L Meredith
- Division of Diabetes, Endocrinology & Gastroenterology; University of Manchester; Manchester United Kingdom
| | - Tiffany-Jayne Allen
- Division of Diabetes, Endocrinology & Gastroenterology; University of Manchester; Manchester United Kingdom
| | - Leanne Walker
- Division of Diabetes, Endocrinology & Gastroenterology; University of Manchester; Manchester United Kingdom
| | - Brian A Telfer
- Division of Pharmacy & Optometry, School of Biology, Medicine and Health; University of Manchester; Manchester United Kingdom
| | | | - Kaye J Williams
- Division of Pharmacy & Optometry, School of Biology, Medicine and Health; University of Manchester; Manchester United Kingdom
| | - Anne White
- Division of Diabetes, Endocrinology & Gastroenterology; University of Manchester; Manchester United Kingdom
| |
Collapse
|
14
|
Gordon RJ, Panigrahi SK, Meece K, Atalayer D, Smiley R, Wardlaw SL. Effects of Opioid Antagonism on Cerebrospinal Fluid Melanocortin Peptides and Cortisol Levels in Humans. J Endocr Soc 2017; 1:1235-1246. [PMID: 29264449 PMCID: PMC5686644 DOI: 10.1210/js.2017-00289] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 08/25/2017] [Indexed: 11/19/2022] Open
Abstract
CONTEXT Hypothalamic proopiomelanocortin (POMC) is processed to α-melanocyte-stimulating hormone, which interacts with the melanocortin antagonist agouti-related protein (AgRP), to regulate energy balance. The POMC-derived opioid peptide β-endorphin (β-EP) also affects feeding behavior via interactions with brain µ-opioid receptors (MORs), including autoinhibitory interactions with MOR expressed by POMC neurons. The opioid antagonist naltrexone (NTX) stimulates POMC neurons in rodents and decreases food intake. OBJECTIVE AND DESIGN The effect of NTX on brain POMC in humans was assessed by measuring POMC peptide concentrations in lumbar cerebrospinal fluid (CSF). AgRP and cortisol levels were also measured because both are inhibited by opioids. In a double-blinded crossover study, 14 healthy subjects were given NTX (50 mg daily) or placebo for either 2 or 7 days. RESULTS CSF β-EP levels increased after 2 and 7 days of NTX treatment; CSF POMC levels did not change, but the β-EP-to-POMC ratio increased. CSF AgRP levels did not change, but plasma AgRP levels tended to increase after NTX (P = 0.06). Cortisol increased in plasma and CSF after NTX treatment; these changes correlated positively with changes in AgRP levels. CONCLUSION Opioid antagonism stimulates POMC peptide release into CSF in humans. The increase in the CSF β-EP-to-POMC ratio could indicate selective release of processed peptides or an effect on POMC processing. Furthermore, AgRP and cortisol stimulation by NTX may mitigate POMC-induced decrease in food intake. It remains to be determined if biomarkers in CSF and plasma could be used to predict responses to pharmacotherapy targeting the melanocortin system.
Collapse
Affiliation(s)
- Rebecca J. Gordon
- Department of Pediatrics, Columbia University College of Physicians & Surgeons, New York, NY 10032
| | - Sunil K. Panigrahi
- Department of Medicine, Columbia University College of Physicians & Surgeons, New York, NY 10032
| | - Kana Meece
- Department of Medicine, Columbia University College of Physicians & Surgeons, New York, NY 10032
| | - Deniz Atalayer
- Department of Medicine, Columbia University College of Physicians & Surgeons, New York, NY 10032
| | - Richard Smiley
- Department of Anesthesiology, Columbia University College of Physicians & Surgeons, New York, NY 10032
| | - Sharon L. Wardlaw
- Department of Medicine, Columbia University College of Physicians & Surgeons, New York, NY 10032
| |
Collapse
|
15
|
Gugliandolo A, Rajan TS, Scionti D, Diomede F, Bramanti P, Mazzon E, Trubiani O. Reprogramming of Oncogene Expression in Gingival Mesenchymal Stem Cells Following Long-Term Culture In Vitro. Cell Reprogram 2017; 19:159-170. [DOI: 10.1089/cell.2016.0056] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
| | | | | | - Francesca Diomede
- Stem Cells and Regenerative Medicine Laboratory, Department of Medical, Oral and Biotechnological Sciences, University “G. d'Annunzio”, Chieti-Pescara, Chieti, Italy
| | | | | | - Oriana Trubiani
- Stem Cells and Regenerative Medicine Laboratory, Department of Medical, Oral and Biotechnological Sciences, University “G. d'Annunzio”, Chieti-Pescara, Chieti, Italy
| |
Collapse
|
16
|
Lin X, Chen W, Wei F, Zhou BP, Hung MC, Xie X. POMC maintains tumor-initiating properties of tumor tissue-derived long-term-cultured breast cancer stem cells. Int J Cancer 2017; 140:2517-2525. [PMID: 28214331 DOI: 10.1002/ijc.30658] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 02/11/2017] [Accepted: 02/13/2017] [Indexed: 12/22/2022]
Abstract
The identification and understanding of the molecular network of cancer stem cells (CSCs) have had a profound impact on our view of carcinogenesis and treatment strategy. Unfortunately, a major problem is that serial passages of CSCs from clinical solid tumor specimens currently are not available in any lab, and thus, reported data are difficult to confirm and intensively interrogated. Here, we have generated two tumor tissue-derived breast CSC (BCSC) lines that showed prolonged maintenance over 20 serial passages in vitro, while retaining their tumor-initiating biological properties. We then deciphered the intrinsic mechanism using analyses of mRNA expression array profiles. It has been determined that pro-opiomelanocortin (POMC) is closely related with protein phosphorylation mediated by G-protein-coupled estrogen receptor (GPER) in BCSC. Following, knockdown of POMC inhibits properties of mammosphere formation, CD44+ CD24- population, CD44 expression, and clonogenicity ability in BCSC. We found that inhibition of POMC attenuates phosphorylation of AKT2 and GSK3β in BCSC. Further in vivo investigations demonstrated that POMC interference regulates proliferation of BCSC-bearing tumors. Combination of the clinical results that POMC positive expression is frequently upregulated in human breast cancer and POMC positivity correlated with a poor prognosis, POMC is a potential therapeutic target for BCSC. In conclusion, we have successfully established two long-term-cultured BCSC from clinical specimens. We further indicated that POMC acts as a potential therapeutic target and prognostic marker for future treatment of BCSC.
Collapse
Affiliation(s)
- Xiaoti Lin
- Department of Breast Oncology, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China.,Department of Oncology, The Affiliated Xiang'an Hospital of Xiamen University, Medical College of Xiamen University, Xiamen, Fujian, 361003, China.,Department of Surgery, Fujian Provincial Tumor Hospital, Fuzhou, 350014, China
| | - Weiyu Chen
- Department of Physiology, Zhongshan medical school, Sun Yat-sen University, Guangzhou, 510060, China
| | - Fengqin Wei
- Department of Oncology, The Affiliated Xiang'an Hospital of Xiamen University, Medical College of Xiamen University, Xiamen, Fujian, 361003, China.,Department of Emergency, Fujian Provincial 2nd People's Hospital, Affiliated Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, 350000, China
| | - Binhua P Zhou
- Department of Breast Oncology, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China.,Department of Molecular and Cellular Biochemistry, Markey Cancer Center, College of Medicine, University of Kentucky, Lexington, KY
| | - Mien-Chie Hung
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Xiaoming Xie
- Department of Breast Oncology, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| |
Collapse
|
17
|
Messaritakis I, Politaki E, Plataki M, Karavassilis V, Kentepozidis N, Koinis F, Samantas E, Georgoulias V, Kotsakis A. Heterogeneity of circulating tumor cells (CTCs) in patients with recurrent small cell lung cancer (SCLC) treated with pazopanib. Lung Cancer 2017; 104:16-23. [DOI: 10.1016/j.lungcan.2016.12.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 12/07/2016] [Accepted: 12/13/2016] [Indexed: 11/28/2022]
|
18
|
Gevers EF, Meredith S, Shah P, Torpiano J, Peters C, Sebire NJ, Slater O, White A, Dattani MT. Cushing syndrome in a child due to pro-opiomelanocortin (POMC) secretion from a yolk sac tumor. Eur J Endocrinol 2017; 176:K1-K7. [PMID: 27879325 DOI: 10.1530/eje-16-0776] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2016] [Revised: 11/14/2016] [Accepted: 11/22/2016] [Indexed: 11/08/2022]
Abstract
CONTEXT Pituitary microadenomas and adrenal tumours are the most common causes for endogenous Cushing syndrome (CS) in children. CASE DESCRIPTION We describe a two-year old girl with Cushing syndrome due to ectopic pro-opiomelanocortin (POMC) production from an abdominal yolk sac tumor. Cortisol concentrations were elevated but adrenocorticotropic hormone (ACTH) concentrations were equivocal. The use of antibodies specifically detecting ACTH precursors revealed that plasma ACTH precursors were elevated. Additionally, an ACTH assay with a low cross-reactivity for precursors showed low concentrations of ACTH. Immunohistochemistry suggested POMC but not ACTH production by the tumour. CONCLUSION We describe a yolk sac tumour as a novel source of ectopic POMC production leading to CS in a young girl.
Collapse
Affiliation(s)
- Evelien F Gevers
- Department of EndocrinologyGreat Ormond Street Hospital for Children, London, UK
| | - Suzanne Meredith
- Division of DiabetesEndocrinology and Gastroenterology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, UK
| | - Pratik Shah
- Department of EndocrinologyGreat Ormond Street Hospital for Children, London, UK
- Section of Genetics and Epigenetics in Health and DiseaseGenetics and Genomic Medicine Programme, UCL Institute of Child Health, London, UK
| | - John Torpiano
- Department of PaediatricsPaediatric Endocrine Service, Mater Dei Hospital, Msida, Malta
| | - Catherine Peters
- Department of EndocrinologyGreat Ormond Street Hospital for Children, London, UK
| | - Neil J Sebire
- Department of HistopathologyGreat Ormond Street Hospital and Institute for Child Health (UCL), London, UK
| | - Olga Slater
- Department of OncologyGreat Ormond Street Hospital for Children, London, UK
| | - Anne White
- Division of DiabetesEndocrinology and Gastroenterology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, UK
| | - Mehul T Dattani
- Department of EndocrinologyGreat Ormond Street Hospital for Children, London, UK
- Section of Genetics and Epigenetics in Health and DiseaseGenetics and Genomic Medicine Programme, UCL Institute of Child Health, London, UK
| |
Collapse
|
19
|
Page-Wilson G, Nguyen KT, Atalayer D, Meece K, Bainbridge HA, Korner J, Gordon RJ, Panigrahi SK, White A, Smiley R, Wardlaw SL. Evaluation of CSF and plasma biomarkers of brain melanocortin activity in response to caloric restriction in humans. Am J Physiol Endocrinol Metab 2017; 312:E19-E26. [PMID: 27894065 PMCID: PMC5283881 DOI: 10.1152/ajpendo.00330.2016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 11/11/2016] [Indexed: 11/22/2022]
Abstract
The melanocortin neuronal system, which consists of hypothalamic proopiomelanocortin (POMC) and agouti-related protein (AgRP) neurons, is a leptin target that regulates energy balance and metabolism, but studies in humans are limited by a lack of reliable biomarkers to assess brain melanocortin activity. The objective of this study was to measure the POMC prohormone and its processed peptide, β-endorphin (β-EP), in cerebrospinal fluid (CSF) and AgRP in CSF and plasma after calorie restriction to validate their utility as biomarkers of brain melanocortin activity. CSF and plasma were obtained from 10 lean and obese subjects after fasting (40 h) and refeeding (24 h), and from 8 obese subjects before and after 6 wk of dieting (800 kcal/day) to assess changes in neuropeptide and hormone levels. After fasting, plasma leptin decreased to 35%, and AgRP increased to 153% of baseline. During refeeding, AgRP declined as leptin increased; CSF β-EP increased, but POMC did not change. Relative changes in plasma and CSF leptin were blunted in obese subjects. After dieting, plasma and CSF leptin decreased to 46% and 70% of baseline, CSF POMC and β-EP decreased, and plasma AgRP increased. At baseline, AgRP correlated negatively with insulin and homeostasis model assessment (HOMA-IR), and positively with the Matsuda index. Thus, following chronic calorie restriction, POMC and β-EP declined in CSF, whereas acutely, only β-EP changed. Plasma AgRP, however, increased after both acute and chronic calorie restriction. These results support the use of CSF POMC and plasma AgRP as biomarkers of hypothalamic melanocortin activity and provide evidence linking AgRP to insulin sensitivity.
Collapse
Affiliation(s)
- Gabrielle Page-Wilson
- Department of Medicine, Columbia University College of Physicians & Surgeons, New York, New York
| | - Kim T Nguyen
- Department of Medicine, Columbia University College of Physicians & Surgeons, New York, New York
| | - Deniz Atalayer
- Department of Medicine, Columbia University College of Physicians & Surgeons, New York, New York
| | - Kana Meece
- Department of Medicine, Columbia University College of Physicians & Surgeons, New York, New York
| | - Heather A Bainbridge
- Department of Medicine, Columbia University College of Physicians & Surgeons, New York, New York
| | - Judith Korner
- Department of Medicine, Columbia University College of Physicians & Surgeons, New York, New York
| | - Rebecca J Gordon
- Department of Pediatrics, Columbia University College of Physicians & Surgeons, New York, New York
| | - Sunil K Panigrahi
- Department of Medicine, Columbia University College of Physicians & Surgeons, New York, New York
| | - Anne White
- Faculties of Life Sciences and Medical and Human Sciences, University of Manchester, Manchester, United Kingdom; and
| | - Richard Smiley
- Department of Anesthesiology, Columbia University College of Physicians & Surgeons, New York, New York
| | - Sharon L Wardlaw
- Department of Medicine, Columbia University College of Physicians & Surgeons, New York, New York;
| |
Collapse
|
20
|
Pore M, Meijer C, de Bock GH, Boersma-van Ek W, Terstappen LWMM, Groen HJM, Timens W, Kruyt FAE, Hiltermann TJN. Cancer Stem Cells, Epithelial to Mesenchymal Markers, and Circulating Tumor Cells in Small Cell Lung Cancer. Clin Lung Cancer 2016; 17:535-542. [PMID: 27363902 DOI: 10.1016/j.cllc.2016.05.015] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 05/26/2016] [Accepted: 05/31/2016] [Indexed: 11/25/2022]
Abstract
BACKGROUND Small cell lung cancer (SCLC) has a poor prognosis, and even with localized (limited) disease, the 5-year survival has only been around 20%. Elevated levels of circulating tumor cells (CTCs) have been associated with a worse prognosis, and markers of cancer stem cells (CSCs) and epithelial to mesenchymal transition have been associated with increased chemoresistance and metastatic spread in SCLC. PATIENTS AND METHODS The biopsy specimens of 38 SCLC patients were used for marker evaluation by immunohistochemistry. The markers for CSCs were CD44 and SOX2. The markers for epithelial to mesenchymal transition were E-cadherin, epithelial cell adhesion molecule, cytokeratins 8, 18, and 19, vimentin, and c-MET. Staining was scored as low (weak) or high (strong) intensity for SOX2, epithelial cell adhesion molecule, cytokeratins 8, 18, and 19, and c-MET and using the immunoreactive score for CD44, E-cadherin, and vimentin, expressed as low or high expression. RESULTS High expression of c-MET (c-METH) and low expression of E-cadherin (E-cadL) showed a trend toward a better prognosis (P = .07 and P = .09, respectively). The combination of c-METH and E-cadL resulted in significantly better survival (P = .007). The tested markers were not associated with CTCs, although a trend was seen for c-METHE-cadL (P = .09) with low CTCs. The CSC markers SOX2 and CD44 were not associated with overall survival in this patient cohort. CONCLUSION SCLC with a mesenchymal-like phenotype (c-METHE-cadL) is associated with longer survival and showed a trend toward lower CTCs.
Collapse
Affiliation(s)
- Milind Pore
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Coby Meijer
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Geertruida H de Bock
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Wytske Boersma-van Ek
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Leon W M M Terstappen
- Department of Medical Cell Biophysics, University of Twente, Enschede, The Netherlands
| | - Harry J M Groen
- Department of Pulmonary Diseases, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Wim Timens
- Department of Pathology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Frank A E Kruyt
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - T Jeroen N Hiltermann
- Department of Pulmonary Diseases, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| |
Collapse
|
21
|
Hurley JD, Akers AT, Friedman JR, Nolan NA, Brown KC, Dasgupta P. Non-pungent long chain capsaicin-analogs arvanil and olvanil display better anti-invasive activity than capsaicin in human small cell lung cancers. Cell Adh Migr 2016; 11:80-97. [PMID: 27196129 DOI: 10.1080/19336918.2016.1187368] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The nutritional compound capsaicin inhibits the invasion of many types of human cancers. The clinical development of capsaicin as an anti-cancer drug is limited due to its unfavorable side effects like burning sensation, stomach cramps, gut pain and nausea. This study compared the anti-invasive activity of capsaicin to non-pungent long chain capsaicin analogs, namely arvanil and olvanil, in human small cell lung cancer cells. Boyden chamber invasion assays revealed that arvanil and olvanil displayed improved anti-invasive activity relative to capsaicin in human SCLC cells. The results of the Boyden chamber assay were confirmed by the spherical invasion assay, and similar results were obtained. The anti-invasive activity of arvanil, olvanil and capsaicin were independent of TRPV and CB1 receptors. Furthermore, the anti-invasive activity of arvanil, olvanil and capsaicin was mediated by the AMPK pathway. Depletion of AMPK levels by siRNA methodology abrogated the anti-invasive activity of arvanil, olvanil and capsaicin. The non-pungent capsaicin analogs arvanil and olvanil display improved anti-invasive activity relative to capsaicin in human SCLC cells. These agents may represent the second generation of capsaicin-like compounds which are more potent than the parent molecule and have a better side effect profile.
Collapse
Affiliation(s)
- John D Hurley
- a Department of Pharmacology, Physiology, and Toxicology , Joan C. Edwards School of Medicine, Marshall University , Huntington , WV , USA
| | - Austin T Akers
- a Department of Pharmacology, Physiology, and Toxicology , Joan C. Edwards School of Medicine, Marshall University , Huntington , WV , USA
| | - Jamie R Friedman
- a Department of Pharmacology, Physiology, and Toxicology , Joan C. Edwards School of Medicine, Marshall University , Huntington , WV , USA
| | - Nicholas A Nolan
- a Department of Pharmacology, Physiology, and Toxicology , Joan C. Edwards School of Medicine, Marshall University , Huntington , WV , USA
| | - Kathleen C Brown
- a Department of Pharmacology, Physiology, and Toxicology , Joan C. Edwards School of Medicine, Marshall University , Huntington , WV , USA
| | - Piyali Dasgupta
- a Department of Pharmacology, Physiology, and Toxicology , Joan C. Edwards School of Medicine, Marshall University , Huntington , WV , USA
| |
Collapse
|
22
|
Meredith SL, Bryant JL, Babur M, Riddell PW, Behrouzi R, Williams KJ, White A. Irradiation Decreases the Neuroendocrine Biomarker Pro-Opiomelanocortin in Small Cell Lung Cancer Cells In Vitro and In Vivo. PLoS One 2016; 11:e0148404. [PMID: 26848743 PMCID: PMC4746075 DOI: 10.1371/journal.pone.0148404] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 01/18/2016] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Small cell lung cancer (SCLC) is an extremely aggressive disease, commonly displaying therapy-resistant relapse. We have previously identified neuroendocrine and epithelial phenotypes in SCLC tumours and the neuroendocrine marker, pro-opiomelanocortin (POMC), correlated with worse overall survival in patients. However, the effect of treatment on these phenotypes is not understood. The current study aimed to determine the effect of repeated irradiation treatment on SCLC cell phenotype, focussing on the neuroendocrine marker, POMC. RESULTS Human SCLC cells (DMS 79) were established as subcutaneous xenograft tumours in CBA nude mice and then exposed to repeated 2Gy irradiation. In untreated animals, POMC in the blood closely mirrored tumour growth; an ideal characteristic for a circulating biomarker. Following repeated localised irradiation in vivo, circulating POMC decreased (p< 0.01), in parallel with a decrease in tumour size, but remained low even when the tumours re-established. The excised tumours displayed reduced and distinctly heterogeneous expression of POMC compared to untreated tumours. There was no difference in the epithelial marker, cytokeratin. However, there were significantly more N-cadherin positive cells in the irradiated tumours. To investigate the tumour response to irradiation, DMS79 cells were repeatedly irradiated in vitro and the surviving cells selected. POMC expression was reduced, while mesenchymal markers N-cadherin, β1-integrin, fibroblast-specific protein 1, β-catenin and Zeb1 expression were amplified in the more irradiation-primed cells. There were no consistent changes in epithelial marker expression. Cell morphology changed dramatically with repeatedly irradiated cells displaying a more elongated shape, suggesting a switch to a more mesenchymal phenotype. CONCLUSIONS In summary, POMC biomarker expression and secretion were reduced in SCLC tumours which regrew after irradiation and in repeatedly irradiation (irradiation-primed) cells. Therefore, POMC was no longer predictive of tumour burden. This highlights the importance of fully evaluating biomarkers during and after therapy to assess clinical utility. Furthermore, the gain in mesenchymal characteristics in irradiated cells could be indicative of a more invasive phenotype.
Collapse
Affiliation(s)
- Suzanne L. Meredith
- Faculty of Medical and Human Sciences, University of Manchester, Manchester, M13 9PT, United Kingdom
| | - Jennifer L. Bryant
- Faculty of Medical and Human Sciences, University of Manchester, Manchester, M13 9PT, United Kingdom
- Faculty of Life Sciences, University of Manchester, Manchester, M13 9PT, United Kingdom
| | - Muhammad Babur
- Faculty of Medical and Human Sciences, University of Manchester, Manchester, M13 9PT, United Kingdom
| | - Philip W. Riddell
- Faculty of Life Sciences, University of Manchester, Manchester, M13 9PT, United Kingdom
| | - Roya Behrouzi
- Faculty of Medical and Human Sciences, University of Manchester, Manchester, M13 9PT, United Kingdom
| | - Kaye J. Williams
- Faculty of Medical and Human Sciences, University of Manchester, Manchester, M13 9PT, United Kingdom
| | - Anne White
- Faculty of Medical and Human Sciences, University of Manchester, Manchester, M13 9PT, United Kingdom
- Faculty of Life Sciences, University of Manchester, Manchester, M13 9PT, United Kingdom
| |
Collapse
|
23
|
Abstract
Small cell lung cancer (SCLC) is an aggressive neuroendocrine tumor of the lung with a tendency to metastasize widely early in the course of the disease. The VA staging system classifies the disease into limited stage (LS) which is confined to one hemithorax and can be included into one radiation field or extensive stage (ES) which extends beyond one hemithorax. Current standard of care is concurrent chemoradiation for LS disease and chemotherapy alone for ES disease. Only a quarter of patients with LS disease will be cured with current standard treatments and majority of the patients ultimately succumb to their disease. A very complex genetic landscape of SCLC accounts for its resistance to conventional therapy and a high recurrence rate, however, at the same time this complexity can form the basis for effective targeted therapy for the disease. In recent years, several different therapeutic strategies and targeted agents have been under investigation for their potential role in SCLC. Several of them including EGFR TKIs, BCR-ABL TKIs, mTOR inhibitors, and VEGF inhibitors have been unsuccessful in showing a survival advantage in this disease. Several others including DNA repair inhibitors, cellular developmental pathway inhibitors, antibody drug conjugates (ADCs), as well as immune therapy with vaccines, immunomodulators, and immune checkpoint inhibitors are being tested. So far, none of these agents are approved for use in SCLC and the majority are in phase I/II clinical trials, with immune checkpoint inhibitors being the most promising therapeutic strategy. In this article, we will discuss these novel therapeutic agents and currently available data in SCLC.
Collapse
Affiliation(s)
- Hirva Mamdani
- 1 Division of Hematology/Oncology, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN, USA ; 2 Levine Cancer Institute, Carolinas HealthCare Systems, Albemarle, NC, USA
| | - Raghava Induru
- 1 Division of Hematology/Oncology, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN, USA ; 2 Levine Cancer Institute, Carolinas HealthCare Systems, Albemarle, NC, USA
| | - Shadia I Jalal
- 1 Division of Hematology/Oncology, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN, USA ; 2 Levine Cancer Institute, Carolinas HealthCare Systems, Albemarle, NC, USA
| |
Collapse
|
24
|
Page-Wilson G, Meece K, White A, Rosenbaum M, Leibel RL, Smiley R, Wardlaw SL. Proopiomelanocortin, agouti-related protein, and leptin in human cerebrospinal fluid: correlations with body weight and adiposity. Am J Physiol Endocrinol Metab 2015; 309:E458-65. [PMID: 26152765 PMCID: PMC4556883 DOI: 10.1152/ajpendo.00206.2015] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 07/01/2015] [Indexed: 11/22/2022]
Abstract
Leptin and its neuronal targets, which produce proopiomelanocortin (POMC) and agouti-related protein (AgRP), regulate energy balance. This study characterized leptin, POMC, and AgRP in the cerebrospinal fluid (CSF) of 47 healthy human subjects, 23 lean and 24 overweight/obese (OW/OB), as related to BMI, adiposity, plasma leptin, soluble leptin receptor (s-OB-R), and insulin. POMC was measured since the POMC prohormone is the predominant POMC peptide in CSF and correlates with hypothalamic POMC in rodents. Plasma AgRP was similarly characterized. CSF leptin was 83-fold lower than in plasma and correlated strongly with BMI, body fat, and insulin. The relative amount of leptin transported into CSF declined with increasing BMI, ranging from 4.5 to 0.52%, consistent with a saturable transport mechanism. CSF sOB-R was 78-fold lower than in plasma and correlated negatively with plasma and CSF leptin. CSF POMC was higher in lean vs. OW/OB subjects (P < 0.001) and correlated negatively with CSF leptin (r = -0.60, P < 0.001) and with plasma leptin, insulin, BMI, and adiposity. CSF AgRP was not different in lean vs. OW/OB; however, plasma AgRP was higher in lean subjects (P = 0.001) and correlated negatively with BMI, adiposity, leptin, insulin, and HOMA (P < 0.005). Thus, CSF measurements may provide useful biomarkers for brain leptin and POMC activity. The striking negative correlation between CSF leptin and POMC could be secondary to leptin resistance and/or neuronal changes associated with obesity but may also indicate that POMC plays a primary role in regulating body weight and adiposity. The role of plasma AgRP as a neuroendocrine biomarker deserves further study.
Collapse
Affiliation(s)
- Gabrielle Page-Wilson
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, New York
| | - Kana Meece
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, New York
| | - Anne White
- Faculties of Life Sciences and Medical and Human Sciences, University of Manchester, Manchester, United Kingdom
| | - Michael Rosenbaum
- Department of Pediatrics, Columbia University College of Physicians and Surgeons, New York, New York; and
| | - Rudolph L Leibel
- Department of Pediatrics, Columbia University College of Physicians and Surgeons, New York, New York; and
| | - Richard Smiley
- Department of Anesthesiology, Columbia University College of Physicians and Surgeons, New York, New York
| | - Sharon L Wardlaw
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, New York;
| |
Collapse
|
25
|
Hao L, Zhao X, Zhang B, Li C, Wang C. Positive expression of pro-opiomelanocortin (POMC) is a novel independent poor prognostic marker in surgically resected non-small cell lung cancer. Tumour Biol 2014; 36:1811-7. [PMID: 25377161 DOI: 10.1007/s13277-014-2784-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Accepted: 10/29/2014] [Indexed: 12/22/2022] Open
Abstract
This study aims to investigate the expression level of pro-opiomelanocortin (POMC) and its prognostic value in non-small cell lung cancer (NSCLC). Immunohistochemical staining was used to detect the expression level of POMC. Correlations between POMC expression and clinical and pathological characteristics were evaluated with the chi-square test, and the prognostic value was determined with the Kaplan-Meier method and COX proportional hazards model, α < 0.05. Of the samples, 48.0% had positive POMC expression. POMC expression was significantly related to poorly differentiated tumors, N-stage, p-stage, postoperative failure pattern, expression of vimentin, and expression of E-cadherin (P < 0.05). Multivariate analysis revealed that POMC-positive expression was an independent risk factor for disease-free survival (hazard ratio (HR) 1.988, 95% confidence interval (CI) 1.094-3.910, P = 0.024) and overall survival (HR 1.892, 95% CI 1.726-3.709, P = 0.036). The addition of POMC protein expression to the prognostic model using pathological stage markedly improved the prognostic potential, and the area under the ROC increased from 0.691 to 0.775. Further study revealed that patients with POMC-negative expression can benefit more from a regimen of paclitaxel and carboplatin chemotherapy than a regimen of vinorelbine and carboplatin compared to patients with POMC-positive expression. We found that POMC-positive expression is a novel, independent poor prognostic marker in patients with NSCLC. Prospective studies are needed to validate the potential prognostic value of POMC in combination with the current staging system and in consideration of adjuvant chemotherapy.
Collapse
Affiliation(s)
- Ligang Hao
- Department of Lung Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Huanhu West Road, Tianjin, 300060, China
| | | | | | | | | |
Collapse
|
26
|
Page-Wilson G, Freda PU, Jacobs TP, Khandji AG, Bruce JN, Foo ST, Meece K, White A, Wardlaw SL. Clinical utility of plasma POMC and AgRP measurements in the differential diagnosis of ACTH-dependent Cushing's syndrome. J Clin Endocrinol Metab 2014; 99:E1838-45. [PMID: 25013995 PMCID: PMC4184073 DOI: 10.1210/jc.2014-1448] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
CONTEXT Distinguishing between pituitary [Cushing's disease (CD)] and ectopic causes [ectopic ACTH syndrome (EAS)] of ACTH-dependent Cushing's syndrome can be challenging. Inferior petrosal sinus sampling (IPSS) best discriminates between CD and occult EAS but is a specialized procedure that is not widely available. Identifying adjunctive diagnostic tests may prove useful. In EAS, abnormal processing of the ACTH precursor proopiomelanocortin (POMC) and the accumulation of POMC-derived peptides might be expected and abnormal levels of other neuropeptides may be detected. OBJECTIVE The objective of the study was to evaluate the diagnostic utility of POMC measurements for distinguishing between CD and occult EAS in patients referred for IPSS. Another objective of the study was to evaluate in parallel the diagnostic utility of another neuropeptide, agouti-related protein (AgRP), because we have observed a 10-fold elevation of AgRP in plasma in a patient with EAS from small-cell lung cancer. DESIGN AND PARTICIPANTS Plasma POMC and AgRP were measured in 38 Cushing's syndrome patients presenting for IPSS, with either no pituitary lesion or a microadenoma on magnetic resonance imaging, and in 38 healthy controls. RESULTS Twenty-seven of 38 patients had CD; 11 of 38 had EAS. The mean POMC was higher in EAS vs CD [54.5 ± 13.0 (SEM) vs 17.2 ± 1.5 fmol/mL; P < .05]. Mean AgRP was higher in EAS vs CD (280 ± 76 vs 120 ± 16 pg/mL; P = .01). Although there was an overlap in POMC and AgRP levels between the groups, the POMC levels greater than 36 fmol/mL (n = 7) and AgRP levels greater than 280 pg/mL (n = 3) were specific for EAS. When used together, POMC greater than 36 fmol/mL and/or AgRP greater than 280 pg/mL detected 9 of 11 cases of EAS, indicating that elevations in these peptides have a high positive predictive value for occult EAS. CONCLUSIONS Expanding upon previous observations of high POMC in EAS, this study specifically demonstrates elevated POMC levels can identify occult ectopic tumors. Elevations in AgRP also favor the diagnosis of EAS, suggesting AgRP should be further evaluated as a potential neuroendocrine tumor marker.
Collapse
Affiliation(s)
- Gabrielle Page-Wilson
- Department of Medicine (G.P.-W., P.U.F., T.P.J., K.M., S.L.W.), Department of Radiology (A.G.K.), Department of Neurological Surgery (J.N.B.), Columbia University College of Physicians & Surgeons, New York, New York 10032; Department of Medicine (S.T.F.), Mt Sinai/St Luke's Roosevelt Hospital, New York, New York 10019; and Faculties of Life Sciences and Medical and Human Sciences (A.W.), University of Manchester, Manchester M13 9PT, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Wardlaw SL, Burant CF, Klein S, Meece K, White A, Kasten T, Lucey BP, Bateman RJ. Continuous 24-hour leptin, proopiomelanocortin, and amino acid measurements in human cerebrospinal fluid: correlations with plasma leptin, soluble leptin receptor, and amino acid levels. J Clin Endocrinol Metab 2014; 99:2540-8. [PMID: 24670082 PMCID: PMC4079306 DOI: 10.1210/jc.2013-4087] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
CONTEXT In order to characterize diurnal changes in central leptin and its target neuropeptide, proopiomelanocortin (POMC), we measured leptin and POMC in cerebrospinal fluid (CSF) as related to changes in plasma leptin and soluble leptin receptor (sOB-R) levels. CSF and plasma levels of 20 amino acids (AA) were also measured because AA can affect brain POMC. DESIGN AND PARTICIPANTS Stored CSF and plasma samples obtained from eight healthy subjects who served as controls for a previous study were evaluated. CSF was collected hourly over 33 h via indwelling subarachnoid catheter. Leptin, sOB-R, and POMC were measured by sensitive ELISA and AA by gas chromatography-mass spectrometry. RESULTS There was a diurnal rhythm for plasma leptin with a peak at 2200 h (144% of baseline) and there was a similar diurnal rhythm for CSF leptin with a peak (117%) 3-5 h after the plasma peak. Plasma sOB-R was lowest at 0300 h and correlated negatively with plasma and CSF leptin. A diurnal rhythm for POMC in CSF was also detected with a peak (125%) at 0100 h. A positive correlation existed between CSF POMC and leptin in individual subjects over time. CSF levels of many AA increased at night. There was a significant correlation between CSF POMC and 10 AA, including leucine, isoleucine, tryptophan, and tyrosine. CONCLUSIONS Diurnal changes occur in leptin and POMC in human CSF that likely reflect changes in central leptin and melanocortin activity. Our results suggest that nocturnal elevations in leptin, AA, and POMC may help to suppress appetite and feeding at night.
Collapse
Affiliation(s)
- Sharon L Wardlaw
- Department of Medicine (S.L.W., K.M.), Columbia University College of Physicians & Surgeons, New York, New York 10032; Department of Internal Medicine (C.F.B.), University of Michigan Medical School, Ann Arbor, Michigan 48019; Center for Human Nutrition and Atkins Center for Excellence in Obesity Medicine (S.K.), Washington University School of Medicine, St Louis, Missouri 63110; Faculties of Life Sciences and Medical and Human Sciences (A.W.), University of Manchester, Manchester M13 9PL, United Kingdom; and Department of Neurology (T.K., B.P.L., R.J.B), Washington University School of Medicine, St Louis, Missouri 63110
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Schlossmacher G, Platt E, Davies A, Meredith S, White A. Glucocorticoid receptor-mediated apoptosis in small-cell lung cancer requires interaction with BCL2. Endocr Relat Cancer 2013; 20:785-95. [PMID: 24036132 DOI: 10.1530/erc-13-0402] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Small-cell lung cancer (SCLC) tumours are highly aggressive. At the time of diagnosis, patients have often developed metastases, and overall prognosis is particularly poor, making effective treatment difficult. Novel mechanisms need to be identified as treatment targets. We have previously found low levels of the glucocorticoid receptor (GR) in SCLC cell lines and demonstrated that over-expression of GR increases tumour cell death both in vitro and in vivo. We hypothesise that low levels of GR impair its inhibitory effect on BCL2 and thus provide a survival advantage to SCLC cell lines. The mechanism behind GR-induced apoptosis is currently unknown; therefore, pro- and anti-apoptotic genes were investigated for their role in GR-mediated apoptosis signalling. We found that over-expression of wtGR via retroviral transduction causes the DMS 79 SCLC cell line to undergo caspase-mediated apoptosis within 72 h. Neither BAD nor BCL2L11 (BIM) mRNA and protein levels were affected by GR restoration implying that GR does not trigger apoptosis in the SCLC cell lines by up-regulating these pro-apoptotic genes. The anti-apoptotic BCL2 gene was significantly overexpressed in six SCLC cell lines and the BCL2 inhibitor ABT-737 increased apoptosis in all three cell lines tested. GR interacted with BCL2 in DMS 153, DMS 79 and COR-L42 cell lines, suggesting that a protein interaction between GR and BCL2 could play a role in GR-induced apoptosis. A deeper understanding of the molecular mechanism for increasing GR expression in SCLC could provide novel treatment strategies in the future.
Collapse
Affiliation(s)
- G Schlossmacher
- Faculty of Life Sciences, Centre for Endocrinology and Diabetes Faculty of Medical and Human Sciences, Manchester Academic Health Sciences Centre, University of Manchester, 3.016 AV Hill Building, Manchester M13 9PT, UK
| | | | | | | | | |
Collapse
|
29
|
Harno E, Cottrell EC, Keevil BG, DeSchoolmeester J, Bohlooly-Y M, Andersén H, Turnbull AV, Leighton B, White A. 11-Dehydrocorticosterone causes metabolic syndrome, which is prevented when 11β-HSD1 is knocked out in livers of male mice. Endocrinology 2013; 154:3599-609. [PMID: 23832962 DOI: 10.1210/en.2013-1362] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Metabolic syndrome is growing in importance with the rising levels of obesity, type 2 diabetes, and insulin resistance. Metabolic syndrome shares many characteristics with Cushing's syndrome, which has led to investigation of the link between excess glucocorticoids and metabolic syndrome. Indeed, increased glucocorticoids from intracellular regeneration by 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) drives insulin resistance and increases adiposity, but these metabolic changes are assumed to be due to increased circulating glucocorticoids. We hypothesized that increasing the substrate for 11β-HSD1 (11-dehydrocorticosterone, 11-DHC) would adversely affect metabolic parameters. We found that chronic administration of 11-DHC to male C57BL/6J mice resulted in increased circulating glucocorticoids, and down-regulation of the hypothalamic-pituitary-adrenal axis. This elevated 11β-HSD1-derived corticosterone led to increased body weight gain and adiposity and produced marked insulin resistance. Surprisingly liver-specific 11β-HSD1 knockout (LKO) mice given 11-DHC did not show any of the adverse metabolic effects seen in wild-type mice. This occurred despite the 11-DHC administration resulting in elevated circulating corticosterone, presumably from adipose tissue. Mice with global deletion of 11β-HSD1 (global knockout) were unaffected by treatment with 11-DHC, having no increase in circulating corticosterone and exhibiting no signs of metabolic impairment. Taken together, these data show that in the absence of 11β-HSD1 in the liver, mice are protected from the metabolic effects of 11-DHC administration, even though circulating glucocorticoids are increased. This implies that liver-derived intratissue glucocorticoids, rather than circulating glucocorticoids, contribute significantly to the development of metabolic syndrome and suggest that local action within hepatic tissue mediates these effects.
Collapse
Affiliation(s)
- Erika Harno
- Faculty of Life Sciences, AV Hill Building, University of Manchester, Manchester, M13 9PT, United Kingdom.
| | | | | | | | | | | | | | | | | |
Collapse
|