1
|
Anand-Ivell R, Heng K, Antonio L, Bartfai G, Casanueva FF, Maggi M, O'Neill TW, Punab M, Rastrelli G, Slowikowska-Hilczer J, Tournoy J, Vanderschueren D, Wu FC, Huhtaniemi IT, Ivell R. Insulin-like peptide 3 (INSL3) as an indicator of leydig cell insufficiency (LCI) in Middle-aged and older men with hypogonadism: reference range and threshold. Aging Male 2024; 27:2346322. [PMID: 38676285 DOI: 10.1080/13685538.2024.2346322] [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: 02/16/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Insulin-like peptide 3 (INSL3) is a circulating biomarker for Leydig cell functional capacity in men, also indicating Leydig Cell Insufficiency (LCI) and potential primary hypogonadism. Using results from large cohort studies we explore sources of biological and technical variance, and establish a reference range for adult men. It is constitutively secreted with little within-individual variation and reflects testicular capacity to produce testosterone. The main INSL3 assays available indicate good concordance with low technical variance; there is no effect of ethnicity. INSL3 declines with age from 35 years at about 15% per decade. Like low calculated free testosterone, and to a lesser extent low total testosterone, reduced INSL3 is significantly associated with increasing age-related morbidity, including lower overall sexual function, reflecting LCI. Consequently, low INSL3 (≤0.4 ng/ml; ca. <2 SD from the population mean) might serve as an additional biochemical marker in the assessment of functional hypogonadism (late-onset hypogonadism, LOH) where testosterone is in the borderline low range. Excluding individuals with low LCI (INSL3 ≤ 0.4 ng/ml) leads to an age-independent (> 35 years) reference range (serum) for INSL3 in the eugonadal population of 0.4 - 2.3 ng/ml, with low INSL3 prospectively identifying individuals at risk of increased future morbidity.
Collapse
Affiliation(s)
| | - Kee Heng
- School of Biosciences, University of Nottingham, Sutton Bonington, UK
| | - Leen Antonio
- Department of Chronic Diseases and Metabolism, Laboratory of Clinical and Experimental Endocrinology, Leuven, KU, Belgium
- Department of Endocrinology, University Hospitals Leuven, Leuven, Belgium
| | - Gyorgy Bartfai
- Department of Obstetrics, Gynaecology and Andrology, Albert Szent-Gyorgy Medical University, Szeged, Hungary
| | - Felipe F Casanueva
- Department of Medicine, Santiago de Compostela University, Complejo Hospitalario Universitario de Santiago (IDIS), CIBER de Fisiopatología Obesidad y Nutricion (CB06/03), Instituto Salud Carlos III, Santiago de Compostela, Spain
| | - Mario Maggi
- Endocrinology and Andrology Unit, "Mario Serio" Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Terence W O'Neill
- Centre for Epidemiology Versus Arthritis, The University of Manchester & NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester, UK
| | - Margus Punab
- Andrology Clinic, Tartu University Hospital, and Institute of Clinical Medicine, and Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Giulia Rastrelli
- Endocrinology and Andrology Unit, "Mario Serio" Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | | | - Jos Tournoy
- Department of Geriatrics, University Hospitals Leuven, and Department of Public Health and Primary Care, KU Leuven, Leuven, Belgium
| | - Dirk Vanderschueren
- Department of Chronic Diseases and Metabolism, Laboratory of Clinical and Experimental Endocrinology, Leuven, KU, Belgium
- Department of Endocrinology, University Hospitals Leuven, Leuven, Belgium
| | - Frederick Cw Wu
- Department of Endocrinology, Manchester University NHS Foundation Trust, Manchester, UK
| | - Ilpo T Huhtaniemi
- Institute of Reproductive and Developmental, Department of Metabolism, Digestion and Reproduction, Imperial College London, Hammersmith Campus, London, UK
| | - Richard Ivell
- School of Biosciences, University of Nottingham, Sutton Bonington, UK
- School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington, UK
| |
Collapse
|
2
|
Kwok SH, Liu Y, Bilder D, Kim J. Paraneoplastic renal dysfunction in fly cancer models driven by inflammatory activation of stem cells. Proc Natl Acad Sci U S A 2024; 121:e2405860121. [PMID: 39392665 PMCID: PMC11494367 DOI: 10.1073/pnas.2405860121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 08/21/2024] [Indexed: 10/12/2024] Open
Abstract
Tumors can induce systemic disturbances in distant organs, leading to physiological changes that enhance host morbidity. In Drosophila cancer models, tumors have been known for decades to cause hypervolemic "bloating" of the abdominal cavity. Here we use allograft and transgenic tumors to show that hosts display fluid retention associated with autonomously defective secretory capacity of fly renal tubules, which function analogous to those of the human kidney. Excretion from these organs is blocked by abnormal cells that originate from inappropriate activation of normally quiescent renal stem cells (RSCs). Blockage is initiated by IL-6-like oncokines that perturb renal water-transporting cells and trigger a damage response in RSCs that proceeds pathologically. Thus, a chronic inflammatory state produced by the tumor causes paraneoplastic fluid dysregulation by altering cellular homeostasis of host renal units.
Collapse
Affiliation(s)
- Sze Hang Kwok
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Yuejiang Liu
- Department of Molecular and Cell Biology, University of California-Berkeley, Berkeley, CA94720
| | - David Bilder
- Department of Molecular and Cell Biology, University of California-Berkeley, Berkeley, CA94720
| | - Jung Kim
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| |
Collapse
|
3
|
Zhang SS, Larrabee L, Chang AH, Desai S, Sloan L, Wang X, Wu Y, Parvez N, Amaratunga K, Hartman AC, Whitnall A, Mason J, Barton NP, Chu AY, Davitte JM, Csakai AJ, Tibbetts CV, Tolbert AE, O'Keefe H, Polanco J, Foley J, Kmett C, Kehler J, Kozejova G, Wang F, Mayer AP, Koenig P, Foletti D, Pitts SJ, Schnackenberg CG. Discovery of RXFP2 genetic association in resistant hypertensive men and RXFP2 antagonists for the treatment of resistant hypertension. Sci Rep 2024; 14:13209. [PMID: 38851835 PMCID: PMC11162469 DOI: 10.1038/s41598-024-62804-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 05/21/2024] [Indexed: 06/10/2024] Open
Abstract
Hypertension remains a leading cause of cardiovascular and kidney diseases. Failure to control blood pressure with ≥ 3 medications or control requiring ≥ 4 medications is classified as resistant hypertension (rHTN) and new therapies are needed to reduce the resulting increased risk of morbidity and mortality. Here, we report genetic evidence that relaxin family peptide receptor 2 (RXFP2) is associated with rHTN in men, but not in women. This study shows that adrenal gland gene expression of RXFP2 is increased in men with hypertension and the RXFP2 natural ligand, INSL3, increases adrenal steroidogenesis and corticosteroid secretion in human adrenal cells. To address the hypothesis that RXFP2 activation is an important mechanism in rHTN, we discovered and characterized small molecule and monoclonal antibody (mAb) blockers of RXFP2. The novel chemical entities and mAbs show potent, selective inhibition of RXFP2 and reduce aldosterone and cortisol synthesis and release. The RXFP2 mAbs have suitable rat pharmacokinetic profiles to evaluate the role of RXFP2 in the development and maintenance of rHTN. Overall, we identified RXFP2 activity as a potential new mechanism in rHTN and discovered RXFP2 antagonists for the future interrogation of RXFP2 in cardiovascular and renal diseases.
Collapse
Affiliation(s)
- Shan-Shan Zhang
- Therapeutics Division, 23andMe, 349 Oyster Point Blvd, South San Francisco, CA, 94080, USA
| | - Lance Larrabee
- Therapeutics Division, 23andMe, 349 Oyster Point Blvd, South San Francisco, CA, 94080, USA
| | - Andrew H Chang
- Therapeutics Division, 23andMe, 349 Oyster Point Blvd, South San Francisco, CA, 94080, USA
| | - Sapna Desai
- Medicinal Science and Technology, GSK, Medicines Research Centre, Gunnels Wood Road, Stevenage, SG1 2NY, UK
| | - Lisa Sloan
- Medicinal Science and Technology, GSK, Medicines Research Centre, Gunnels Wood Road, Stevenage, SG1 2NY, UK
| | - Xin Wang
- Research, 23andMe, 223 N Mathilda Ave., Sunnyvale, CA, 94086, USA
| | - Yixuan Wu
- Therapeutics Division, 23andMe, 349 Oyster Point Blvd, South San Francisco, CA, 94080, USA
| | - Nazia Parvez
- Medicinal Science and Technology, GSK, Medicines Research Centre, Gunnels Wood Road, Stevenage, SG1 2NY, UK
| | - Karen Amaratunga
- Medicinal Science and Technology, GSK, Medicines Research Centre, Gunnels Wood Road, Stevenage, SG1 2NY, UK
| | - Allison C Hartman
- Medicinal Science and Technology, GSK, 1250 S. Collegeville Rd., Collegeville, PA, 19426, USA
| | - Abby Whitnall
- Medicinal Science and Technology, GSK, Medicines Research Centre, Gunnels Wood Road, Stevenage, SG1 2NY, UK
| | - Joseph Mason
- Medicinal Science and Technology, GSK, Medicines Research Centre, Gunnels Wood Road, Stevenage, SG1 2NY, UK
| | - Nicholas P Barton
- Medicinal Science and Technology, GSK, Medicines Research Centre, Gunnels Wood Road, Stevenage, SG1 2NY, UK
| | - Audrey Y Chu
- Genomic Sciences, GSK, 300 Technology Square, Cambridge, MA, 02139, USA
| | | | - Adam J Csakai
- Medicinal Science and Technology, GSK, 200 Cambridgepark Drive, Cambridge, MA, 02140, USA
| | | | - Audrey E Tolbert
- Medicinal Science and Technology, GSK, 200 Cambridgepark Drive, Cambridge, MA, 02140, USA
| | - Heather O'Keefe
- Medicinal Science and Technology, GSK, 200 Cambridgepark Drive, Cambridge, MA, 02140, USA
| | - Jessie Polanco
- Therapeutics Division, 23andMe, 349 Oyster Point Blvd, South San Francisco, CA, 94080, USA
| | - Joseph Foley
- Novel Human Genetics Research Unit, GSK, 1250 S. Collegeville Rd., Collegeville, PA, 19426, USA
| | - Casey Kmett
- DMPK, GSK, 1250 S. Collegeville Rd, Collegeville, PA, 19426, USA
| | - Jonathan Kehler
- Bioanalysis, Immunogenicity and Biomarkers, GSK, 1250 S. Collegeville Rd., Collegeville, PA, 19426, USA
| | - Gabriela Kozejova
- Medicinal Science and Technology, GSK, Medicines Research Centre, Gunnels Wood Road, Stevenage, SG1 2NY, UK
| | - Feng Wang
- DMPK, GSK, 1250 S. Collegeville Rd, Collegeville, PA, 19426, USA
| | - Andrew P Mayer
- Bioanalysis, Immunogenicity and Biomarkers, GSK, 1250 S. Collegeville Rd., Collegeville, PA, 19426, USA
| | - Patrick Koenig
- Therapeutics Division, 23andMe, 349 Oyster Point Blvd, South San Francisco, CA, 94080, USA
| | - Davide Foletti
- Therapeutics Division, 23andMe, 349 Oyster Point Blvd, South San Francisco, CA, 94080, USA
| | - Steven J Pitts
- Research, 23andMe, 223 N Mathilda Ave., Sunnyvale, CA, 94086, USA
| | | |
Collapse
|
4
|
Yoon JW, Baek SE, Yang JY, Yeom E. NUCB1 is required for proper insulin signaling to control longevity in Drosophila. Geriatr Gerontol Int 2024; 24:486-492. [PMID: 38509017 DOI: 10.1111/ggi.14858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 02/09/2024] [Accepted: 02/27/2024] [Indexed: 03/22/2024]
Abstract
AIM We examined the novel role of NUCB1(Nucleobindin-1) associated with longevity in Drosophila melanogaster. METHODS We measured the lifespan, metabolic phenotypes, and mRNA levels of Drosophila insulin-like peptides (Dilps), the protein level of phosphorylated AKT, and the localization of FOXO and its target gene expressions in the NUCB1 knockdown condition. RESULTS NUCB1 knockdown flies show an extended lifespan and metabolic phenotypes such as increased circulating glucose level and starvation resistance. The mRNA expression levels of Dilps and the protein level of phosphorylated AKT, a downstream component of insulin signaling, were decreased in NUCB1 knockdown flies compared with the control flies. Also, the nuclear localization of FOXO and its target gene expressions, such as d4E-BP and InR, were elevated. CONCLUSIONS The results show that NUCB1 knockdown flies exhibits an extended lifespan. These findings suggest that NUCB1 modulates longevity through insulin signaling in Drosophila. Geriatr Gerontol Int 2024; 24: 486-492.
Collapse
Affiliation(s)
- Jong-Won Yoon
- School of Life Science and Biotechnology, College of Natural Sciences, Kyungpook National University, Daegu, Korea
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, Korea
| | - Si-Eun Baek
- School of Life Science and Biotechnology, College of Natural Sciences, Kyungpook National University, Daegu, Korea
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, Korea
| | - Jae-Yoon Yang
- School of Life Science and Biotechnology, College of Natural Sciences, Kyungpook National University, Daegu, Korea
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, Korea
| | - Eunbyul Yeom
- School of Life Science and Biotechnology, College of Natural Sciences, Kyungpook National University, Daegu, Korea
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, Korea
| |
Collapse
|
5
|
Qiu S, Wu Q, Wang H, Liu D, Chen C, Zhu Z, Zheng H, Yang G, Li L, Yang M. AZGP1 in POMC neurons modulates energy homeostasis and metabolism through leptin-mediated STAT3 phosphorylation. Nat Commun 2024; 15:3377. [PMID: 38643150 PMCID: PMC11032411 DOI: 10.1038/s41467-024-47684-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 04/10/2024] [Indexed: 04/22/2024] Open
Abstract
Zinc-alpha2-glycoprotein (AZGP1) has been implicated in peripheral metabolism; however, its role in regulating energy metabolism in the brain, particularly in POMC neurons, remains unknown. Here, we show that AZGP1 in POMC neurons plays a crucial role in controlling whole-body metabolism. POMC neuron-specific overexpression of Azgp1 under high-fat diet conditions reduces energy intake, raises energy expenditure, elevates peripheral tissue leptin and insulin sensitivity, alleviates liver steatosis, and promotes adipose tissue browning. Conversely, mice with inducible deletion of Azgp1 in POMC neurons exhibit the opposite metabolic phenotypes, showing increased susceptibility to diet-induced obesity. Notably, an increase in AZGP1 signaling in the hypothalamus elevates STAT3 phosphorylation and increases POMC neuron excitability. Mechanistically, AZGP1 enhances leptin-JAK2-STAT3 signaling by interacting with acylglycerol kinase (AGK) to block its ubiquitination degradation. Collectively, these results suggest that AZGP1 plays a crucial role in regulating energy homeostasis and glucose/lipid metabolism by acting on hypothalamic POMC neurons.
Collapse
Affiliation(s)
- Sheng Qiu
- Department of Endocrinology, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, China
- Key Laboratory of Medical Diagnostics of Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Qinan Wu
- Department of Endocrinology, The Affiliated Dazu Hospital of Chongqing Medical University, Chongqing, 402360, China
| | - Hao Wang
- Department of Endocrinology, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, China
| | - Dongfang Liu
- Department of Endocrinology, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, China
| | - Chen Chen
- Endocrinology, SBMS, Faculty of Medicine, University of Queensland, Brisbane, QLD, 4072, Australia
| | - Zhiming Zhu
- Department of Hypertension and Endocrinology, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Hongting Zheng
- Department of Endocrinology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Gangyi Yang
- Department of Endocrinology, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, China.
| | - Ling Li
- Department of Endocrinology, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, China.
- Key Laboratory of Medical Diagnostics of Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China.
| | - Mengliu Yang
- Department of Endocrinology, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, China.
| |
Collapse
|
6
|
Kwok SH, Liu Y, Bilder D, Kim J. Paraneoplastic renal dysfunction in fly cancer models driven by inflammatory activation of stem cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.21.586173. [PMID: 38585959 PMCID: PMC10996499 DOI: 10.1101/2024.03.21.586173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Tumors can induce systemic disturbances in distant organs, leading to physiological changes that enhance host morbidity. In Drosophila cancer models, tumors have been known for decades to cause hypervolemic 'bloating' of the abdominal cavity. Here we use allograft and transgenic tumors to show that hosts display fluid retention associated with autonomously defective secretory capacity of fly renal tubules, which function analogous to those of the human kidney. Excretion from these organs is blocked by abnormal cells that originate from inappropriate activation of normally quiescent renal stem cells (RSCs). Blockage is initiated by IL-6-like oncokines that perturb renal water-transporting cells, and trigger a damage response in RSCs that proceeds pathologically. Thus, a chronic inflammatory state produced by the tumor causes paraneoplastic fluid dysregulation by altering cellular homeostasis of host renal units.
Collapse
Affiliation(s)
- Sze Hang Kwok
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Yuejiang Liu
- Department of Molecular and Cell Biology, University of California-Berkeley, Berkeley CA, 94720, USA
| | - David Bilder
- Department of Molecular and Cell Biology, University of California-Berkeley, Berkeley CA, 94720, USA
| | - Jung Kim
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Department of Molecular and Cell Biology, University of California-Berkeley, Berkeley CA, 94720, USA
| |
Collapse
|
7
|
Ispoglou T, McCullough D, Windle A, Nair S, Cox N, White H, Burke D, Kanatas A, Prokopidis K. Addressing cancer anorexia-cachexia in older patients: Potential therapeutic strategies and molecular pathways. Clin Nutr 2024; 43:552-566. [PMID: 38237369 DOI: 10.1016/j.clnu.2024.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 01/04/2024] [Accepted: 01/10/2024] [Indexed: 02/03/2024]
Abstract
Cancer cachexia (CC) syndrome, a feature of cancer-associated muscle wasting, is particularly pronounced in older patients, and is characterised by decreased energy intake and upregulated skeletal muscle catabolic pathways. To address CC, appetite stimulants, anabolic drugs, cytokine mediators, essential amino acid supplementation, nutritional counselling, cognitive behavioural therapy, and enteral nutrition have been utilised. However, pharmacological treatments that have also shown promising results, such as megestrol acetate, anamorelin, thalidomide, and delta-9-tetrahydrocannabinol, have been associated with gastrointestinal and cardiovascular complications. Emerging evidence on the efficacy of probiotics in modulating gut microbiota also presents a promising adjunct to traditional therapies, potentially enhancing nutritional absorption and systemic inflammation control. Additionally, low-dose olanzapine has demonstrated improved appetite and weight management in older patients undergoing chemotherapy, offering a potential refinement to current therapeutic approaches. This review aims to elucidate the molecular mechanisms underpinning CC, with a particular focus on the role of anorexia in exacerbating muscle wasting, and to propose pharmacological and non-pharmacological strategies to mitigate this syndrome, particularly emphasising the needs of an older demographic. Future research targeting CC should focus on refining appetite-stimulating drugs with fewer side-effects, specifically catering to the needs of older patients, and investigating nutritional factors that can either enhance appetite or minimise suppression of appetite in individuals with CC, especially within this vulnerable group.
Collapse
Affiliation(s)
| | | | - Angela Windle
- Department of Nursing and Midwifery, School of Human and Health Sciences, University of Huddersfield, Huddersfield, UK; School of Medicine, University of Leeds, Leeds, UK
| | | | - Natalie Cox
- Academic Geriatric Medicine, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Helen White
- School of Health, Leeds Beckett University, Leeds, UK
| | - Dermot Burke
- School of Medicine, University of Leeds, Leeds, UK
| | | | - Konstantinos Prokopidis
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK; Liverpool Centre for Cardiovascular Science, University of Liverpool, Liverpool, UK
| |
Collapse
|
8
|
Jung J, Han H. The diverse influences of relaxin-like peptide family on tumor progression: Potential opportunities and emerging challenges. Heliyon 2024; 10:e24463. [PMID: 38298643 PMCID: PMC10828710 DOI: 10.1016/j.heliyon.2024.e24463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 01/05/2024] [Accepted: 01/09/2024] [Indexed: 02/02/2024] Open
Abstract
Relaxin-like peptide family exhibit differential expression patterns in various types of cancers and play a role in cancer development. This family participates in tumorigenic processes encompassing proliferation, migration, invasion, tumor microenvironment, immune microenvironment, and anti-cancer resistance, ultimately influencing patient prognosis. In this review, we explore the mechanisms underlying the interaction between the RLN-like peptide family and tumors and provide an overview of therapeutic approaches utilizing this interaction.
Collapse
Affiliation(s)
| | - Hyunho Han
- Department of Urology, Urological Science Institute, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| |
Collapse
|
9
|
Wang Y, Dong Z, An Z, Jin W. Cancer cachexia: Focus on cachexia factors and inter-organ communication. Chin Med J (Engl) 2024; 137:44-62. [PMID: 37968131 PMCID: PMC10766315 DOI: 10.1097/cm9.0000000000002846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Indexed: 11/17/2023] Open
Abstract
ABSTRACT Cancer cachexia is a multi-organ syndrome and closely related to changes in signal communication between organs, which is mediated by cancer cachexia factors. Cancer cachexia factors, being the general name of inflammatory factors, circulating proteins, metabolites, and microRNA secreted by tumor or host cells, play a role in secretory or other organs and mediate complex signal communication between organs during cancer cachexia. Cancer cachexia factors are also a potential target for the diagnosis and treatment. The pathogenesis of cachexia is unclear and no clear effective treatment is available. Thus, the treatment of cancer cachexia from the perspective of the tumor ecosystem rather than from the perspective of a single molecule and a single organ is urgently needed. From the point of signal communication between organs mediated by cancer cachexia factors, finding a deeper understanding of the pathogenesis, diagnosis, and treatment of cancer cachexia is of great significance to improve the level of diagnosis and treatment. This review begins with cancer cachexia factors released during the interaction between tumor and host cells, and provides a comprehensive summary of the pathogenesis, diagnosis, and treatment for cancer cachexia, along with a particular sight on multi-organ signal communication mediated by cancer cachexia factors. This summary aims to deepen medical community's understanding of cancer cachexia and may conduce to the discovery of new diagnostic and therapeutic targets for cancer cachexia.
Collapse
Affiliation(s)
- Yongfei Wang
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu 730000, China
- Institute of Cancer Neuroscience, Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu 730000, China
| | - Zikai Dong
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu 730000, China
- Institute of Cancer Neuroscience, Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu 730000, China
| | - Ziyi An
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu 730000, China
- Institute of Cancer Neuroscience, Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu 730000, China
| | - Weilin Jin
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu 730000, China
- Institute of Cancer Neuroscience, Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu 730000, China
| |
Collapse
|
10
|
Xu W, Li G, Chen Y, Ye X, Song W. A novel antidiuretic hormone governs tumour-induced renal dysfunction. Nature 2023; 624:425-432. [PMID: 38057665 DOI: 10.1038/s41586-023-06833-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Accepted: 11/03/2023] [Indexed: 12/08/2023]
Abstract
Maintenance of renal function and fluid transport are essential for vertebrates and invertebrates to adapt to physiological and pathological challenges. Human patients with malignant tumours frequently develop detrimental renal dysfunction and oliguria, and previous studies suggest the involvement of chemotherapeutic toxicity and tumour-associated inflammation1,2. However, how tumours might directly modulate renal functions remains largely unclear. Here, using conserved tumour models in Drosophila melanogaster3, we characterized isoform F of ion transport peptide (ITPF) as a fly antidiuretic hormone that is secreted by a subset of yki3SA gut tumour cells, impairs renal function and causes severe abdomen bloating and fluid accumulation. Mechanistically, tumour-derived ITPF targets the G-protein-coupled receptor TkR99D in stellate cells of Malpighian tubules-an excretory organ that is equivalent to renal tubules4-to activate nitric oxide synthase-cGMP signalling and inhibit fluid excretion. We further uncovered antidiuretic functions of mammalian neurokinin 3 receptor (NK3R), the homologue of fly TkR99D, as pharmaceutical blockade of NK3R efficiently alleviates renal tubular dysfunction in mice bearing different malignant tumours. Together, our results demonstrate a novel antidiuretic pathway mediating tumour-renal crosstalk across species and offer therapeutic opportunities for the treatment of cancer-associated renal dysfunction.
Collapse
Affiliation(s)
- Wenhao Xu
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, Hubei, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Gerui Li
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, Hubei, China
| | - Yuan Chen
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, Hubei, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Xujun Ye
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, Hubei, China.
| | - Wei Song
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, Hubei, China.
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, China.
| |
Collapse
|
11
|
Pranoto IKA, Lee J, Kwon YV. The roles of the native cell differentiation program aberrantly recapitulated in Drosophila intestinal tumors. Cell Rep 2023; 42:113245. [PMID: 37837622 PMCID: PMC10872463 DOI: 10.1016/j.celrep.2023.113245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 06/11/2023] [Accepted: 09/26/2023] [Indexed: 10/16/2023] Open
Abstract
Many tumors recapitulate the developmental and differentiation program of their tissue of origin, a basis for tumor cell heterogeneity. Although stem-cell-like tumor cells are well studied, the roles of tumor cells undergoing differentiation remain to be elucidated. We employ Drosophila genetics to demonstrate that the differentiation program of intestinal stem cells is crucial for enabling intestinal tumors to invade and induce non-tumor-autonomous phenotypes. The differentiation program that generates absorptive cells is aberrantly recapitulated in the intestinal tumors generated by activation of the Yap1 ortholog Yorkie. Inhibiting it allows stem-cell-like tumor cells to grow but suppresses invasiveness and reshapes various phenotypes associated with cachexia-like wasting by altering the expression of tumor-derived factors. Our study provides insight into how a native differentiation program determines a tumor's capacity to induce advanced cancer phenotypes and suggests that manipulating the differentiation programs co-opted in tumors might alleviate complications of cancer, including cachexia.
Collapse
Affiliation(s)
| | - Jiae Lee
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Young V Kwon
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
12
|
González Segarra AJ, Pontes G, Jourjine N, Del Toro A, Scott K. Hunger- and thirst-sensing neurons modulate a neuroendocrine network to coordinate sugar and water ingestion. eLife 2023; 12:RP88143. [PMID: 37732734 PMCID: PMC10513480 DOI: 10.7554/elife.88143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2023] Open
Abstract
Consumption of food and water is tightly regulated by the nervous system to maintain internal nutrient homeostasis. Although generally considered independently, interactions between hunger and thirst drives are important to coordinate competing needs. In Drosophila, four neurons called the interoceptive subesophageal zone neurons (ISNs) respond to intrinsic hunger and thirst signals to oppositely regulate sucrose and water ingestion. Here, we investigate the neural circuit downstream of the ISNs to examine how ingestion is regulated based on internal needs. Utilizing the recently available fly brain connectome, we find that the ISNs synapse with a novel cell-type bilateral T-shaped neuron (BiT) that projects to neuroendocrine centers. In vivo neural manipulations revealed that BiT oppositely regulates sugar and water ingestion. Neuroendocrine cells downstream of ISNs include several peptide-releasing and peptide-sensing neurons, including insulin producing cells (IPCs), crustacean cardioactive peptide (CCAP) neurons, and CCHamide-2 receptor isoform RA (CCHa2R-RA) neurons. These neurons contribute differentially to ingestion of sugar and water, with IPCs and CCAP neurons oppositely regulating sugar and water ingestion, and CCHa2R-RA neurons modulating only water ingestion. Thus, the decision to consume sugar or water occurs via regulation of a broad peptidergic network that integrates internal signals of nutritional state to generate nutrient-specific ingestion.
Collapse
Affiliation(s)
| | - Gina Pontes
- University of California, BerkeleyBerkeleyUnited States
| | | | | | - Kristin Scott
- University of California, BerkeleyBerkeleyUnited States
| |
Collapse
|
13
|
Winter LM, Reinhardt D, Schatter A, Tissen V, Wiora H, Gerlach D, Tontsch-Grunt U, Colbatzky F, Stierstorfer B, Yun SW. Molecular basis of GDF15 induction and suppression by drugs in cardiomyocytes and cancer cells toward precision medicine. Sci Rep 2023; 13:12061. [PMID: 37495707 PMCID: PMC10372009 DOI: 10.1038/s41598-023-38450-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 07/08/2023] [Indexed: 07/28/2023] Open
Abstract
GDF15 has recently emerged as a key driver of the development of various disease conditions including cancer cachexia. Not only the tumor itself but also adverse effects of chemotherapy have been reported to contribute to increased GDF15. Although regulation of GDF15 transcription by BET domain has recently been reported, the molecular mechanisms of GDF15 gene regulation by drugs are still unknown, leaving uncertainty about the safe and effective therapeutic strategies targeting GDF15. We screened various cardiotoxic drugs and BET inhibitors for their effects on GDF15 regulation in human cardiomyocytes and cancer cell lines and analyzed in-house and public gene signature databases. We found that DNA damaging drugs induce GDF15 in cardiomyocytes more strongly than drugs with other modes of action. In cancer cells, GDF15 induction varied depending on drug- and cell type-specific gene signatures including mutations in PI3KCA, TP53, BRAF and MUC16. GDF15 suppression by BET inhibition is particularly effective in cancer cells with low activity of the PI3K/Akt axis and high extracellular concentrations of pantothenate. Our findings provide insights that the risk for GDF15 overexpression and concomitant cachexia can be reduced by a personalized selection of anticancer drugs and patients for precision medicine.
Collapse
Affiliation(s)
- Lisa-Maria Winter
- Boehringer Ingelheim Pharma GmbH & Co KG, Birkendorfer Strasse 65, 88397, Biberach an Der Riß, Germany
| | - Diana Reinhardt
- Boehringer Ingelheim Pharma GmbH & Co KG, Birkendorfer Strasse 65, 88397, Biberach an Der Riß, Germany
| | - Ariane Schatter
- Boehringer Ingelheim Pharma GmbH & Co KG, Birkendorfer Strasse 65, 88397, Biberach an Der Riß, Germany
| | - Vivien Tissen
- Boehringer Ingelheim Pharma GmbH & Co KG, Birkendorfer Strasse 65, 88397, Biberach an Der Riß, Germany
| | - Heike Wiora
- Boehringer Ingelheim Pharma GmbH & Co KG, Birkendorfer Strasse 65, 88397, Biberach an Der Riß, Germany
| | - Daniel Gerlach
- Boehringer Ingelheim RCV, GmbH & Co KG, 1120, Vienna, Austria
| | | | - Florian Colbatzky
- Boehringer Ingelheim Pharma GmbH & Co KG, Birkendorfer Strasse 65, 88397, Biberach an Der Riß, Germany
| | - Birgit Stierstorfer
- Boehringer Ingelheim Pharma GmbH & Co KG, Birkendorfer Strasse 65, 88397, Biberach an Der Riß, Germany
| | - Seong-Wook Yun
- Boehringer Ingelheim Pharma GmbH & Co KG, Birkendorfer Strasse 65, 88397, Biberach an Der Riß, Germany.
| |
Collapse
|
14
|
González-Segarra AJ, Pontes G, Jourjine N, Del Toro A, Scott K. Hunger- and thirst-sensing neurons modulate a neuroendocrine network to coordinate sugar and water ingestion. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.06.535891. [PMID: 37066363 PMCID: PMC10104137 DOI: 10.1101/2023.04.06.535891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
Consumption of food and water is tightly regulated by the nervous system to maintain internal nutrient homeostasis. Although generally considered independently, interactions between hunger and thirst drives are important to coordinate competing needs. In Drosophila , four neurons called the Interoceptive Subesophageal zone Neurons (ISNs) respond to intrinsic hunger and thirst signals to oppositely regulate sucrose and water ingestion. Here, we investigate the neural circuit downstream of the ISNs to examine how ingestion is regulated based on internal needs. Utilizing the recently available fly brain connectome, we find that the ISNs synapse with a novel cell type Bilateral T-shaped neuron (BiT) that projects to neuroendocrine centers. In vivo neural manipulations revealed that BiT oppositely regulates sugar and water ingestion. Neuroendocrine cells downstream of ISNs include several peptide-releasing and peptide-sensing neurons, including insulin producing cells (IPC), crustacean cardioactive peptide (CCAP) neurons, and CCHamide-2 receptor isoform RA (CCHa2R-RA) neurons. These neurons contribute differentially to ingestion of sugar and water, with IPCs and CCAP neurons oppositely regulating sugar and water ingestion, and CCHa2R-RA neurons modulating only water ingestion. Thus, the decision to consume sugar or water occurs via regulation of a broad peptidergic network that integrates internal signals of nutritional state to generate nutrient-specific ingestion.
Collapse
Affiliation(s)
| | - Gina Pontes
- University of California, Berkeley, United States
- present address: IBBEA, CONICET-UBA, Buenos Aires, Argentina
| | - Nicholas Jourjine
- University of California, Berkeley, United States
- present address: Harvard University, Cambridge, United States
| | - Alexander Del Toro
- University of California, Berkeley, United States
- present address: Brown University, Rhode Island, United States
| | | |
Collapse
|
15
|
Laturney M, Sterne GR, Scott K. Mating activates neuroendocrine pathways signaling hunger in Drosophila females. eLife 2023; 12:e85117. [PMID: 37184218 PMCID: PMC10229122 DOI: 10.7554/elife.85117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 05/13/2023] [Indexed: 05/16/2023] Open
Abstract
Mated females reallocate resources to offspring production, causing changes to nutritional requirements and challenges to energy homeostasis. Although observed across species, the neural and endocrine mechanisms that regulate the nutritional needs of mated females are not well understood. Here, we find that mated Drosophila melanogaster females increase sugar intake, which is regulated by the activity of sexually dimorphic insulin receptor (Lgr3) neurons. In virgins, Lgr3+ cells have reduced activity as they receive inhibitory input from active, female-specific pCd-2 cells, restricting sugar intake. During copulation, males deposit sex peptide into the female reproductive tract, which silences a three-tier mating status circuit and initiates the female postmating response. We show that pCd-2 neurons also become silenced after mating due to the direct synaptic input from the mating status circuit. Thus, in mated females pCd-2 inhibition is attenuated, activating downstream Lgr3+ neurons and promoting sugar intake. Together, this circuit transforms the mated signal into a long-term hunger signal. Our results demonstrate that the mating circuit alters nutrient sensing centers to increase feeding in mated females, providing a mechanism to increase intake in anticipation of the energetic costs associated with reproduction.
Collapse
Affiliation(s)
| | | | - Kristin Scott
- University of California, BerkeleyBerkeleyUnited States
| |
Collapse
|
16
|
Ferrer M, Anthony TG, Ayres JS, Biffi G, Brown JC, Caan BJ, Cespedes Feliciano EM, Coll AP, Dunne RF, Goncalves MD, Grethlein J, Heymsfield SB, Hui S, Jamal-Hanjani M, Lam JM, Lewis DY, McCandlish D, Mustian KM, O'Rahilly S, Perrimon N, White EP, Janowitz T. Cachexia: A systemic consequence of progressive, unresolved disease. Cell 2023; 186:1824-1845. [PMID: 37116469 PMCID: PMC11059056 DOI: 10.1016/j.cell.2023.03.028] [Citation(s) in RCA: 47] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 01/15/2023] [Accepted: 03/23/2023] [Indexed: 04/30/2023]
Abstract
Cachexia, a systemic wasting condition, is considered a late consequence of diseases, including cancer, organ failure, or infections, and contributes to significant morbidity and mortality. The induction process and mechanistic progression of cachexia are incompletely understood. Refocusing academic efforts away from advanced cachexia to the etiology of cachexia may enable discoveries of new therapeutic approaches. Here, we review drivers, mechanisms, organismal predispositions, evidence for multi-organ interaction, model systems, clinical research, trials, and care provision from early onset to late cachexia. Evidence is emerging that distinct inflammatory, metabolic, and neuro-modulatory drivers can initiate processes that ultimately converge on advanced cachexia.
Collapse
Affiliation(s)
- Miriam Ferrer
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA; MRC Cancer Unit, University of Cambridge, Hutchison Research Centre, Cambridge Biomedical Campus, Cambridge CB2 0XZ, UK
| | - Tracy G Anthony
- Department of Nutritional Sciences, Rutgers School of Environmental and Biological Sciences, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Janelle S Ayres
- Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Giulia Biffi
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Cambridge CB2 0RE, UK
| | - Justin C Brown
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808, USA
| | - Bette J Caan
- Kaiser Permanente Northern California Division of Research, Oakland, CA 94612, USA
| | | | - Anthony P Coll
- Wellcome Trust-MRC Institute of Metabolic Science and MRC Metabolic Diseases Unit, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Richard F Dunne
- University of Rochester Medical Center, University of Rochester, Rochester, NY 14642, USA
| | - Marcus D Goncalves
- Division of Endocrinology, Department of Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Jonas Grethlein
- Ruprecht Karl University of Heidelberg, Heidelberg 69117, Germany
| | - Steven B Heymsfield
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808, USA
| | - Sheng Hui
- Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA 02115, USA
| | - Mariam Jamal-Hanjani
- Department of Medical Oncology, University College London Hospitals, London WC1E 6DD, UK; Cancer Research UK Lung Cancer Centre of Excellence and Cancer Metastasis Laboratory, University College London Cancer Institute, London WC1E 6DD, UK
| | - Jie Min Lam
- Cancer Research UK Lung Cancer Centre of Excellence and Cancer Metastasis Laboratory, University College London Cancer Institute, London WC1E 6DD, UK
| | - David Y Lewis
- The Beatson Institute for Cancer Research, Cancer Research UK, Glasgow G61 1BD, UK
| | - David McCandlish
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Karen M Mustian
- University of Rochester Medical Center, University of Rochester, Rochester, NY 14642, USA
| | - Stephen O'Rahilly
- Wellcome Trust-MRC Institute of Metabolic Science and MRC Metabolic Diseases Unit, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Norbert Perrimon
- Department of Genetics, Blavatnik Institute, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Eileen P White
- Rutgers Cancer Institute of New Jersey, Department of Molecular Biology and Biochemistry, Rutgers University, The State University of New Jersey, New Brunswick, NJ 08901, USA; Ludwig Princeton Branch, Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ 08544, USA
| | - Tobias Janowitz
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA; Northwell Health Cancer Institute, Northwell Health, New Hyde Park, NY 11042, USA.
| |
Collapse
|
17
|
Raskov H, Gaggar S, Tajik A, Orhan A, Gögenur I. Metabolic switch in cancer - Survival of the fittest. Eur J Cancer 2023; 180:30-51. [PMID: 36527974 DOI: 10.1016/j.ejca.2022.11.025] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 11/21/2022] [Indexed: 11/27/2022]
Abstract
Cell metabolism is characterised by the highly coordinated conversion of nutrients into energy and biomass. In solid cancers, hypoxia, nutrient deficiencies, and tumour vasculature are incompatible with accelerated anabolic growth and require a rewiring of cancer cell metabolism. Driver gene mutations direct malignant cells away from oxidation to maximise energy production and biosynthesis while tumour-secreted factors degrade peripheral tissues to fuel disease progression and initiate metastasis. As it is vital to understand cancer cell metabolism and survival mechanisms, this review discusses the metabolic switch and current drug targets and clinical trials. In the future, metabolic markers may be included when phenotyping individual tumours to improve the therapeutic opportunities for personalised therapy.
Collapse
Affiliation(s)
- Hans Raskov
- Center for Surgical Science, Zealand University Hospital, Køge, 4600, Denmark.
| | - Shruti Gaggar
- Center for Surgical Science, Zealand University Hospital, Køge, 4600, Denmark
| | - Asma Tajik
- Center for Surgical Science, Zealand University Hospital, Køge, 4600, Denmark
| | - Adile Orhan
- Center for Surgical Science, Zealand University Hospital, Køge, 4600, Denmark; Department of Clinical Oncology, Zealand University Hospital, Roskilde, 4000, Denmark
| | - Ismail Gögenur
- Center for Surgical Science, Zealand University Hospital, Køge, 4600, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, 2200, Denmark
| |
Collapse
|
18
|
Li H, Luo X, Li N, Liu T, Zhang J. Insulin-like peptide 8 (Ilp8) regulates female fecundity in flies. Front Cell Dev Biol 2023; 11:1103923. [PMID: 36743416 PMCID: PMC9890075 DOI: 10.3389/fcell.2023.1103923] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 01/04/2023] [Indexed: 01/19/2023] Open
Abstract
Introduction: Insulin-like peptides (Ilps) play crucial roles in nearly all life stages of insects. Ilp8 is involved in developmental stability, stress resistance and female fecundity in several insect species, but the underlying mechanisms are not fully understood. Here we report the functional characterization of Ilp8s in three fly species, including Bactrocera dorsalis, Drosophila mercatorum and Drosophila melanogaster. Methods: Phylogenetic analyses were performed to identify and characterize insect Ilp8s. The amino acid sequences of fly Ilp8s were aligned and the three-dimensional structures of fly Ilp8s were constructed and compared. The tissue specific expression pattern of fly Ilp8s were examined by qRT-PCR. In Bactrocera dorsalis and Drosophila mercatorum, dsRNAs were injected into virgin females to inhibit the expression of Ilp8 and the impacts on female fecundity were examined. In Drosophila melanogaster, the female fecundity of Ilp8 loss-of-function mutant was compared with wild type control flies. The mutant fruit fly strain was also used for sexual behavioral analysis and transcriptomic analysis. Results: Orthologs of Ilp8s are found in major groups of insects except for the lepidopterans and coleopterans, and Ilp8s are found to be well separated from other Ilps in three fly species. The key motif and the predicted three-dimensional structure of fly Ilp8s are well conserved. Ilp8 are specifically expressed in the ovary and are essential for female fecundity in three fly species. Behavior analysis demonstrates that Ilp8 mutation impairs female sexual attractiveness in fruit fly, which results in decreased mating success and is likely the cause of fecundity reduction. Further transcriptomic analysis indicates that Ilp8 might influence metabolism, immune activity, oocyte development as well as hormone homeostasis to collectively regulate female fecundity in the fruit fly. Discussion: Our findings support a universal role of insect Ilp8 in female fecundity, and also provide novel clues for understanding the modes of action of Ilp8.
Collapse
Affiliation(s)
- Haomiao Li
- MARA Key Laboratory of Surveillance and Management for Plant Quarantine Pests, College of Plant Protection, China Agricultural University, Beijing, China
| | - Xi Luo
- MARA Key Laboratory of Surveillance and Management for Plant Quarantine Pests, College of Plant Protection, China Agricultural University, Beijing, China
| | - Na Li
- MARA Key Laboratory of Surveillance and Management for Plant Quarantine Pests, College of Plant Protection, China Agricultural University, Beijing, China
| | - Tao Liu
- Chinese Academy of Inspection and Quarantine, Beijing, China
| | - Junzheng Zhang
- MARA Key Laboratory of Surveillance and Management for Plant Quarantine Pests, College of Plant Protection, China Agricultural University, Beijing, China,*Correspondence: Junzheng Zhang,
| |
Collapse
|
19
|
Li X, Holtrop T, Jansen FAC, Olson B, Levasseur P, Zhu X, Poland M, Schalwijk W, Witkamp RF, Marks DL, van Norren K. Lipopolysaccharide-induced hypothalamic inflammation in cancer cachexia-anorexia is amplified by tumour-derived prostaglandin E2. J Cachexia Sarcopenia Muscle 2022; 13:3014-3027. [PMID: 36303458 PMCID: PMC9745464 DOI: 10.1002/jcsm.13093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 08/17/2022] [Accepted: 09/02/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Cachexia-anorexia syndrome is a complex metabolic condition characterized by skeletal muscle wasting, reduced food intake and prominent involvement of systemic and central inflammation. Here, the gut barrier function was investigated in pancreatic cancer-induced cachexia mouse models by relating intestinal permeability to the degree of cachexia. We further investigated the involvement of the gut-brain axis and the crosstalk between tumour, gut and hypothalamus in vitro. METHODS Two distinct mouse models of pancreatic cancer cachexia (KPC and 4662) were used. Intestinal inflammation and permeability were assessed through fluorescein isothiocyanate dextran (FITC-dextran) and lipopolysaccharide (LPS), and hypothalamic and systemic inflammation through mRNA expression and plasma cytokines, respectively. To simulate the tumour-gut-brain crosstalk, hypothalamic (HypoE-N46) cells were incubated with cachexia-inducing tumour secretomes and LPS. A synthetic mimic of C26 secretome was produced based on its secreted inflammatory mediators. Each component of the mimic was systematically omitted to narrow down the key mediator(s) with an amplifying inflammation. To substantiate its contribution, cyclooxygenase-2 (COX-2) inhibitor was used. RESULTS In vivo experiments showed FITC-dextran was enhanced in the KPC group (362.3 vs. sham 111.4 ng/mL, P < 0.001). LPS was increased to 140.9 ng/mL in the KPC group, compared with sham and 4662 groups (115.8 and 115.8 ng/mL, P < 0.05). Hypothalamic inflammatory gene expression of Ccl2 was up-regulated in the KPC group (6.3 vs. sham 1, P < 0.0001, 4662 1.3, P < 0.001), which significantly correlated with LPS concentration (r = 0.4948, P = 0.0226). These data suggest that intestinal permeability is positively related to the cachexic degree. Prostaglandin E2 (PGE2) was confirmed to be present in the plasma and PGE2 concentration (log10) in the KPC group was much higher than in 4662 group (1.85 and 0.56 ng/mL, P < 0.001), indicating a role for PGE2 in pancreatic cancer-induced cachexia. Parallel to in vivo findings, in vitro experiments revealed that the cachexia-inducing tumour secretomes (C26, LLC, KPC and 4662) amplified LPS-induced hypothalamic IL-6 secretion (419%, 321%, 294%, 160%). COX-2 inhibitor to the tumour cells reduced PGE2 content (from 105 to 102 pg/mL) in the secretomes and eliminated the amplified hypothalamic IL-6 production. Moreover, results could be reproduced by addition of PGE2 alone, indicating that the increased hypothalamic inflammation is directly related to the PGE2 from tumour. CONCLUSIONS PGE2 secreted by the tumour may play a role in amplifying the effects of bacteria-derived LPS on the inflammatory hypothalamic response. The cachexia-inducing potential of tumour mice models parallels the loss of intestinal barrier function. Tumour-derived PGE2 might play a key role in cancer-related cachexia-anorexia syndrome via tumour-gut-brain crosstalk.
Collapse
Affiliation(s)
- Xiaolin Li
- Nutritional Biology, Division of Human Nutrition, Wageningen University, Wageningen, The Netherlands
| | - Tosca Holtrop
- Nutritional Biology, Division of Human Nutrition, Wageningen University, Wageningen, The Netherlands.,Papé Family Pediatric Research Institute, Oregon Health & Science University (OHSU), Portland, OR, USA
| | - Fleur A C Jansen
- Nutritional Biology, Division of Human Nutrition, Wageningen University, Wageningen, The Netherlands
| | - Brennan Olson
- Papé Family Pediatric Research Institute, Oregon Health & Science University (OHSU), Portland, OR, USA
| | - Pete Levasseur
- Papé Family Pediatric Research Institute, Oregon Health & Science University (OHSU), Portland, OR, USA
| | - Xinxia Zhu
- Papé Family Pediatric Research Institute, Oregon Health & Science University (OHSU), Portland, OR, USA
| | - Mieke Poland
- Nutritional Biology, Division of Human Nutrition, Wageningen University, Wageningen, The Netherlands
| | - Winni Schalwijk
- Nutritional Biology, Division of Human Nutrition, Wageningen University, Wageningen, The Netherlands
| | - Renger F Witkamp
- Nutritional Biology, Division of Human Nutrition, Wageningen University, Wageningen, The Netherlands
| | - Daniel L Marks
- Papé Family Pediatric Research Institute, Oregon Health & Science University (OHSU), Portland, OR, USA
| | - Klaske van Norren
- Nutritional Biology, Division of Human Nutrition, Wageningen University, Wageningen, The Netherlands
| |
Collapse
|
20
|
Suh GSB, Yu K, Kim YJ, Oh Y, Park JJ. History of Drosophila neurogenetic research in South Korea. J Neurogenet 2022:1-7. [PMID: 36165786 DOI: 10.1080/01677063.2022.2115040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Abstract
Neurogenetic research using the Drosophila model has immensely expanded around the world. Likewise, scientists in South Korea have leveraged the advantages of Drosophila genetic tools to understand various neurobiological processes. In this special issue, we will overview the history of Drosophila neurogenetic research in South Korea that led to significant discoveries and notably implications. We will describe how Drosophila system was first introduced to elevate neural developmental studies in 1990s. Establishing Drosophila-related resources has been a key venture, which led to the generation of over 100,000 mutant lines and the launch of the K-Gut initiative with Korea Drosophila Research Center (KDRC). These resources have supported the pioneer studies in modeling human disease and understanding genes and neural circuits that regulate animal behavior and physiology.
Collapse
Affiliation(s)
- Greg S B Suh
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Kweon Yu
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Metabolism and Neurophysiology Research Group, Daejeon, Republic of Korea
| | - Young-Joon Kim
- Department of Biological Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| | - Yangkyun Oh
- Department of Life Sciences, Ewha Womans University, Seoul, Republic of Korea
| | - Joong-Jean Park
- Department of Physiology, Korea University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
21
|
Ferrara M, Samaden M, Ruggieri E, Vénéreau E. Cancer cachexia as a multiorgan failure: Reconstruction of the crime scene. Front Cell Dev Biol 2022; 10:960341. [PMID: 36158184 PMCID: PMC9493094 DOI: 10.3389/fcell.2022.960341] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 08/03/2022] [Indexed: 11/13/2022] Open
Abstract
Cachexia is a devastating syndrome associated with the end-stage of several diseases, including cancer, and characterized by body weight loss and severe muscle and adipose tissue wasting. Although different cancer types are affected to diverse extents by cachexia, about 80% of all cancer patients experience this comorbidity, which highly reduces quality of life and response to therapy, and worsens prognosis, accounting for more than 25% of all cancer deaths. Cachexia represents an urgent medical need because, despite several molecular mechanisms have been identified, no effective therapy is currently available for this devastating syndrome. Most studies focus on skeletal muscle, which is indeed the main affected and clinically relevant organ, but cancer cachexia is characterized by a multiorgan failure. In this review, we focus on the current knowledge on the multiple tissues affected by cachexia and on the biomarkers with the attempt to define a chronological pathway, which might be useful for the early identification of patients who will undergo cachexia. Indeed, it is likely that the inefficiency of current therapies might be attributed, at least in part, to their administration in patients at the late stages of cachexia.
Collapse
Affiliation(s)
- Michele Ferrara
- Tissue Regeneration and Homeostasis Unit, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Maria Samaden
- Tissue Regeneration and Homeostasis Unit, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Elena Ruggieri
- Tissue Regeneration and Homeostasis Unit, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Emilie Vénéreau
- Tissue Regeneration and Homeostasis Unit, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
22
|
Transforming Growth Factor-Beta Signaling in Cancer-Induced Cachexia: From Molecular Pathways to the Clinics. Cells 2022; 11:cells11172671. [PMID: 36078078 PMCID: PMC9454487 DOI: 10.3390/cells11172671] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 08/23/2022] [Accepted: 08/25/2022] [Indexed: 02/06/2023] Open
Abstract
Cachexia is a metabolic syndrome consisting of massive loss of muscle mass and function that has a severe impact on the quality of life and survival of cancer patients. Up to 20% of lung cancer patients and up to 80% of pancreatic cancer patients are diagnosed with cachexia, leading to death in 20% of them. The main drivers of cachexia are cytokines such as interleukin-6 (IL-6), tumor necrosis factor-alpha (TNF-α), macrophage inhibitory cytokine 1 (MIC-1/GDF15) and transforming growth factor-beta (TGF-β). Besides its double-edged role as a tumor suppressor and activator, TGF-β causes muscle loss through myostatin-based signaling, involved in the reduction in protein synthesis and enhanced protein degradation. Additionally, TGF-β induces inhibin and activin, causing weight loss and muscle depletion, while MIC-1/GDF15, a member of the TGF-β superfamily, leads to anorexia and so, indirectly, to muscle wasting, acting on the hypothalamus center. Against this background, the blockade of TGF-β is tested as a potential mechanism to revert cachexia, and antibodies against TGF-β reduced weight and muscle loss in murine models of pancreatic cancer. This article reviews the role of the TGF-β pathway and to a minor extent of other molecules including microRNA in cancer onset and progression with a special focus on their involvement in cachexia, to enlighten whether TGF-β and such other players could be potential targets for therapy.
Collapse
|
23
|
Chen Y, Xu W, Chen Y, Han A, Song J, Zhou X, Song W. Renal NF-κB activation impairs uric acid homeostasis to promote tumor-associated mortality independent of wasting. Immunity 2022; 55:1594-1608.e6. [PMID: 36029766 DOI: 10.1016/j.immuni.2022.07.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 05/27/2022] [Accepted: 07/29/2022] [Indexed: 12/12/2022]
Abstract
Tumor-induced host wasting and mortality are general phenomena across species. Many groups have previously demonstrated endocrinal impacts of malignant tumors on host wasting in rodents and Drosophila. Whether and how environmental factors and host immune response contribute to tumor-associated host wasting and survival, however, are largely unknown. Here, we report that flies bearing malignant yki3SA-gut tumors exhibited the exponential increase of commensal bacteria, which were mostly acquired from the environment, and systemic IMD-NF-κB activation due to suppression of a gut antibacterial amidase PGRP-SC2. Either gut microbial elimination or specific IMD-NF-κB blockade in the renal-like Malpighian tubules potently improved mortality of yki3SA-tumor-bearing flies in a manner independent of host wasting. We further indicate that renal IMD-NF-κB activation caused uric acid (UA) overload to reduce survival of tumor-bearing flies. Therefore, our results uncover a fundamental mechanism whereby gut commensal dysbiosis, renal immune activation, and UA imbalance potentiate tumor-associated host death.
Collapse
Affiliation(s)
- Yuchen Chen
- Department of Hepatobiliary and Pancreatic Surgery, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei 430071, China; TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei 430071, China
| | - Wenhao Xu
- Department of Hepatobiliary and Pancreatic Surgery, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei 430071, China; TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei 430071, China
| | - Yuan Chen
- Department of Hepatobiliary and Pancreatic Surgery, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei 430071, China; TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei 430071, China
| | - Anxuan Han
- Department of Hepatobiliary and Pancreatic Surgery, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei 430071, China; TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei 430071, China
| | - Jiantao Song
- Department of Hepatobiliary and Pancreatic Surgery, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei 430071, China; TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei 430071, China
| | - Xiaoya Zhou
- Department of Hepatobiliary and Pancreatic Surgery, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei 430071, China; TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei 430071, China
| | - Wei Song
- Department of Hepatobiliary and Pancreatic Surgery, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei 430071, China; TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei 430071, China.
| |
Collapse
|
24
|
Nässel DR, Zandawala M. Endocrine cybernetics: neuropeptides as molecular switches in behavioural decisions. Open Biol 2022; 12:220174. [PMID: 35892199 PMCID: PMC9326288 DOI: 10.1098/rsob.220174] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Plasticity in animal behaviour relies on the ability to integrate external and internal cues from the changing environment and hence modulate activity in synaptic circuits of the brain. This context-dependent neuromodulation is largely based on non-synaptic signalling with neuropeptides. Here, we describe select peptidergic systems in the Drosophila brain that act at different levels of a hierarchy to modulate behaviour and associated physiology. These systems modulate circuits in brain regions, such as the central complex and the mushroom bodies, which supervise specific behaviours. At the top level of the hierarchy there are small numbers of large peptidergic neurons that arborize widely in multiple areas of the brain to orchestrate or modulate global activity in a state and context-dependent manner. At the bottom level local peptidergic neurons provide executive neuromodulation of sensory gain and intrinsically in restricted parts of specific neuronal circuits. The orchestrating neurons receive interoceptive signals that mediate energy and sleep homeostasis, metabolic state and circadian timing, as well as external cues that affect food search, aggression or mating. Some of these cues can be triggers of conflicting behaviours such as mating versus aggression, or sleep versus feeding, and peptidergic neurons participate in circuits, enabling behaviour choices and switches.
Collapse
Affiliation(s)
- Dick R. Nässel
- Department of Zoology, Stockholm University, 10691 Stockholm, Sweden
| | - Meet Zandawala
- Neurobiology and Genetics, Theodor-Boveri-Institute, Biocenter, University of Würzburg, Am Hubland Würzburg 97074, Germany
| |
Collapse
|
25
|
Understanding the molecular basis of anorexia and tissue wasting in cancer cachexia. Exp Mol Med 2022; 54:426-432. [PMID: 35388147 PMCID: PMC9076846 DOI: 10.1038/s12276-022-00752-w] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 12/15/2021] [Accepted: 01/16/2022] [Indexed: 02/07/2023] Open
Abstract
Cancer cachexia syndrome is a major cause of morbidity and mortality in cancer patients in the advanced stage. It is a devastating disorder characterized by nutritional impairment, weakness, and wasting, and it affects treatment success and quality of life. Two major symptoms of cancer cachexia are anorexia and weight loss. Weight loss in cachexia is not reversed through increased food intake, suggesting that anorexia and weight loss in cancer patients are regulated by independent molecular mechanisms. Although the wasting phenotype mostly occurs in skeletal muscle and adipose tissue, other organs, such as the brain, liver, pancreas, heart, and gut, are also involved in cachexia. Thus, cachexia is a multiorgan syndrome. Although the molecular basis of cancer cachexia-induced weight loss is known, the mechanism underlying anorexia is poorly understood. Here, we highlight our recent discovery of a new anorexia mechanism by which a tumor-derived humoral factor induces cancer anorexia by regulating feeding-related neuropeptide hormones in the brain. Furthermore, we elucidated the process through which anorexia precedes tissue wasting in cachexia. This review article aims to provide an overview of the key molecular mechanisms of anorexia and tissue wasting caused by cancer cachexia. Tumors can release factors that cause anorexia and weight loss in cancer patients, negatively impacting quality of life and treatment success. Patients with this condition, known as cachexia, can lose their appetite and be unable to gain weight even if they eat more. Although cancer cachexia directly causes the death of up to 20% of cancer patients, the mechanisms are poorly understood. Eunbyul Yeom and Kweon Yu at The Korea Research Institute of Bioscience and Biotechnology, Daejon, South Korea have reviewed the causes of cancer cachexia, highlighting their recent discovery that tumors produce a signaling molecule that induces anorexia by disrupting hunger signaling in the brain. Improving our understanding of the mechanisms underlying cancer cachexia may help in development of treatments.
Collapse
|
26
|
Nässel DR, Wu SF. Cholecystokinin/sulfakinin peptide signaling: conserved roles at the intersection between feeding, mating and aggression. Cell Mol Life Sci 2022; 79:188. [PMID: 35286508 PMCID: PMC8921109 DOI: 10.1007/s00018-022-04214-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 02/19/2022] [Accepted: 02/21/2022] [Indexed: 12/27/2022]
Abstract
Neuropeptides are the most diverse messenger molecules in metazoans and are involved in regulation of daily physiology and a wide array of behaviors. Some neuropeptides and their cognate receptors are structurally and functionally well conserved over evolution in bilaterian animals. Among these are peptides related to gastrin and cholecystokinin (CCK). In mammals, CCK is produced by intestinal endocrine cells and brain neurons, and regulates gall bladder contractions, pancreatic enzyme secretion, gut functions, satiety and food intake. Additionally, CCK plays important roles in neuromodulation in several brain circuits that regulate reward, anxiety, aggression and sexual behavior. In invertebrates, CCK-type peptides (sulfakinins, SKs) are, with a few exceptions, produced by brain neurons only. Common among invertebrates is that SKs mediate satiety and regulate food ingestion by a variety of mechanisms. Also regulation of secretion of digestive enzymes has been reported. Studies of the genetically tractable fly Drosophila have advanced our understanding of SK signaling mechanisms in regulation of satiety and feeding, but also in gustatory sensitivity, locomotor activity, aggression and reproductive behavior. A set of eight SK-expressing brain neurons plays important roles in regulation of these competing behaviors. In males, they integrate internal state and external stimuli to diminish sex drive and increase aggression. The same neurons also diminish sugar gustation, induce satiety and reduce feeding. Although several functional roles of CCK/SK signaling appear conserved between Drosophila and mammals, available data suggest that the underlying mechanisms differ.
Collapse
Affiliation(s)
- Dick R Nässel
- Department of Zoology, Stockholm University, 10691, Stockholm, Sweden.
| | - Shun-Fan Wu
- College of Plant Protection/Laboratory of Bio-Interactions and Crop Health, Nanjing Agricultural University, Nanjing, 210095, China
| |
Collapse
|
27
|
Abstract
Cachexia, a wasting syndrome that is often associated with cancer, is one of the primary causes of death in cancer patients. Cancer cachexia occurs largely due to systemic metabolic alterations stimulated by tumors. Despite the prevalence of cachexia, our understanding of how tumors interact with host tissues and how they affect metabolism is limited. Among the challenges of studying tumor-host tissue crosstalk are the complexity of cancer itself and our insufficient knowledge of the factors that tumors release into the blood. Drosophila is emerging as a powerful model in which to identify tumor-derived factors that influence systemic metabolism and tissue wasting. Strikingly, studies that are characterizing factors derived from different fly tumor cachexia models are identifying both common and distinct cachectic molecules, suggesting that cachexia is more than one disease and that fly models can help identify these differences. Here, we review what has been learned from studies of tumor-induced organ wasting in Drosophila and discuss the open questions.
Collapse
Affiliation(s)
- Ying Liu
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Pedro Saavedra
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Norbert Perrimon
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Howard Hughes Medical Institute, Boston, MA 02115, USA
| |
Collapse
|
28
|
Lv S, Qian Z, Li J, Piao S, Li J. Identification and Validation of a Hypoxia-Immune-Based Prognostic mRNA Signature for Oral Squamous Cell Carcinoma. JOURNAL OF ONCOLOGY 2022; 2022:5286251. [PMID: 35178089 PMCID: PMC8844353 DOI: 10.1155/2022/5286251] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 11/22/2021] [Accepted: 12/20/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND Oral squamous cell carcinoma (OSCC) is a commonly encountered head and neck malignancy. Increasing evidence shows that there are abnormal immune response and chronic cell hypoxia in the development of OSCC. However, there is a lack of a reliable hypoxia-immune-based gene signature that may serve to accurately prognosticate OSCC. METHODS The mRNA expression data of OSCC patients were extracted from the TCGA and GEO databases. Hypoxia status was identified using the t-distributed Stochastic Neighbor Embedding (t-SNE) algorithm. Both ESTIMATE and single-sample gene-set enrichment analysis (ssGSEA) were used for further evaluation of immune status. The DEGs in different hypoxia and immune status were determined, and univariate Cox regression was used to identify significantly prognostic genes. A machine learning method, least absolute shrinkage and selection operator (LASSO) Cox regression analysis, allowed us to construct prognostic gene signature to predict the overall survival (OS) of OSCC patients. RESULTS A total of 773 DEGs were identified between hypoxia high and low groups. According to immune cell infiltration, patients were divided into immune high, medium, and low groups and immune-associated DEGs were identified. A total of 193 overlapped DEGs in both immune and hypoxia status were identified. With the univariate and LASSO Cox regression model, eight signature mRNAs (FAM122C, RNF157, RANBP17, SOWAHA, KIAA1211, RIPPLY2, INSL3, and DNAH1) were selected for further calculation of their respective risk scores. The risk score showed a significant association with age and perineural and lymphovascular invasion. In the GEO validation cohort, a better OS was observed in patients from the low-risk group in comparison with those in the high-risk group. High-risk patients also demonstrated different immune infiltration characteristics from the low-risk group and the low-risk group showed potentially better immunotherapy efficacy in contrast to high-risk ones. CONCLUSION The hypoxia-immune-based gene signature has prognostic potential in OSCC.
Collapse
Affiliation(s)
- Shaohua Lv
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150081, China
- Stomatology School, Harbin Medical University, 143 Yiman Street, Nangang District, Harbin, Heilongjiang, China
| | - Zhipeng Qian
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Jianhao Li
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150081, China
| | - Songlin Piao
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150081, China
| | - Jichen Li
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150081, China
- Stomatology School, Harbin Medical University, 143 Yiman Street, Nangang District, Harbin, Heilongjiang, China
| |
Collapse
|
29
|
Yang L, Wang J, Gong X, Fan Q, Yang X, Cui Y, Gao X, Li L, Sun X, Li Y, Wang Y. Emerging Roles for LGR4 in Organ Development, Energy Metabolism and Carcinogenesis. Front Genet 2022; 12:728827. [PMID: 35140734 PMCID: PMC8819683 DOI: 10.3389/fgene.2021.728827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 12/30/2021] [Indexed: 11/26/2022] Open
Abstract
The leucine-rich repeats containing G protein-coupled receptor 4 (LGR4) belonging to G protein-coupled receptors (GPCRs) family, had various regulatory roles at multiple cellular types and numerous targeting sites, and aberrant LGR4 signaling played crucial roles in diseases and carcinogenesis. On the basis of these facts, LGR4 may become an appealing therapeutic target for the treatment of diseases and tumors. However, a comprehensive investigation of its functions and applications was still lacking. Hence, this paper provided an overview of the molecular characteristics and signaling mechanisms of LGR4, its involvement in multiple organ development and participation in the modulation of immunology related diseases, metabolic diseases, and oxidative stress damage along with cancer progression. Given that GPCRs accounted for almost a third of current clinical drug targets, the in-depth understanding of the sophisticated connections of LGR4 and its ligands would not only enrich their regulatory networks, but also shed new light on designing novel molecular targeted drugs and small molecule blockers for revolutionizing the treatment of various diseases and tumors.
Collapse
Affiliation(s)
- Linlin Yang
- Department of Gynecological Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Municipal Key Clinical Specialty, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Disease, Shanghai, China
| | - Jing Wang
- Department of Gynecological Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Municipal Key Clinical Specialty, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Disease, Shanghai, China
| | - Xiaodi Gong
- Department of Gynecological Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Municipal Key Clinical Specialty, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Disease, Shanghai, China
| | - Qiong Fan
- Department of Gynecological Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Municipal Key Clinical Specialty, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Disease, Shanghai, China
| | - Xiaoming Yang
- Department of Gynecological Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Municipal Key Clinical Specialty, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Disease, Shanghai, China
| | - Yunxia Cui
- Department of Gynecological Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Municipal Key Clinical Specialty, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Disease, Shanghai, China
| | - Xiaoyan Gao
- Department of Gynecological Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Municipal Key Clinical Specialty, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Disease, Shanghai, China
| | - Lijuan Li
- Department of Gynecological Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Municipal Key Clinical Specialty, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Disease, Shanghai, China
| | - Xiao Sun
- Department of Gynecological Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Municipal Key Clinical Specialty, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Disease, Shanghai, China
| | - Yuhong Li
- Department of Gynecological Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Municipal Key Clinical Specialty, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Disease, Shanghai, China
- *Correspondence: Yuhong Li, ; Yudong Wang,
| | - Yudong Wang
- Department of Gynecological Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Municipal Key Clinical Specialty, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Disease, Shanghai, China
- *Correspondence: Yuhong Li, ; Yudong Wang,
| |
Collapse
|
30
|
A Blueprint for Cancer-Related Inflammation and Host Innate Immunity. Cells 2021; 10:cells10113211. [PMID: 34831432 PMCID: PMC8623541 DOI: 10.3390/cells10113211] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/27/2021] [Accepted: 11/10/2021] [Indexed: 12/30/2022] Open
Abstract
Both in situ and allograft models of cancer in juvenile and adult Drosophila melanogaster fruit flies offer a powerful means for unravelling cancer gene networks and cancer-host interactions. They can also be used as tools for cost-effective drug discovery and repurposing. Moreover, in situ modeling of emerging tumors makes it possible to address cancer initiating events-a black box in cancer research, tackle the innate antitumor immune responses to incipient preneoplastic cells and recurrent growing tumors, and decipher the initiation and evolution of inflammation. These studies in Drosophila melanogaster can serve as a blueprint for studies in more complex organisms and help in the design of mechanism-based therapies for the individualized treatment of cancer diseases in humans. This review focuses on new discoveries in Drosophila related to the diverse innate immune responses to cancer-related inflammation and the systemic effects that are so detrimental to the host.
Collapse
|
31
|
Bilder D, Ong K, Hsi TC, Adiga K, Kim J. Tumour-host interactions through the lens of Drosophila. Nat Rev Cancer 2021; 21:687-700. [PMID: 34389815 PMCID: PMC8669834 DOI: 10.1038/s41568-021-00387-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/01/2021] [Indexed: 02/07/2023]
Abstract
There is a large gap between the deep understanding of mechanisms driving tumour growth and the reasons why patients ultimately die of cancer. It is now appreciated that interactions between the tumour and surrounding non-tumour (sometimes referred to as host) cells play critical roles in mortality as well as tumour progression, but much remains unknown about the underlying molecular mechanisms, especially those that act beyond the tumour microenvironment. Drosophila has a track record of high-impact discoveries about cell-autonomous growth regulation, and is well suited to now probe mysteries of tumour - host interactions. Here, we review current knowledge about how fly tumours interact with microenvironmental stroma, circulating innate immune cells and distant organs to influence disease progression. We also discuss reciprocal regulation between tumours and host physiology, with a particular focus on paraneoplasias. The fly's simplicity along with the ability to study lethality directly provide an opportunity to shed new light on how cancer actually kills.
Collapse
Affiliation(s)
- David Bilder
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA.
| | - Katy Ong
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Tsai-Ching Hsi
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Kavya Adiga
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Jung Kim
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| |
Collapse
|
32
|
Kim J, Chuang HC, Wolf NK, Nicolai CJ, Raulet DH, Saijo K, Bilder D. Tumor-induced disruption of the blood-brain barrier promotes host death. Dev Cell 2021; 56:2712-2721.e4. [PMID: 34496290 DOI: 10.1016/j.devcel.2021.08.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/29/2021] [Accepted: 08/12/2021] [Indexed: 11/24/2022]
Abstract
Cancer patients often die from symptoms that manifest at a distance from any tumor. Mechanisms underlying these systemic physiological perturbations, called paraneoplastic syndromes, may benefit from investigation in non-mammalian systems. Using a non-metastatic Drosophila adult model, we find that malignant-tumor-produced cytokines drive widespread host activation of JAK-STAT signaling and cause premature lethality. STAT activity is particularly high in cells of the blood-brain barrier (BBB), where it induces aberrant BBB permeability. Remarkably, inhibiting STAT in the BBB not only rescues barrier function but also extends the lifespan of tumor-bearing hosts. We identify BBB damage in other pathological conditions that cause elevated inflammatory signaling, including obesity and infection, where BBB permeability also regulates host survival. IL-6-dependent BBB dysfunction is further seen in a mouse tumor model, and it again promotes host morbidity. Therefore, BBB alterations constitute a conserved lethal tumor-host interaction that also underlies other physiological morbidities.
Collapse
Affiliation(s)
- Jung Kim
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA
| | - Hsiu-Chun Chuang
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA
| | - Natalie K Wolf
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA
| | - Christopher J Nicolai
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA
| | - David H Raulet
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA
| | - Kaoru Saijo
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA; Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA 94720, USA
| | - David Bilder
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
33
|
Olson B, Diba P, Korzun T, Marks DL. Neural Mechanisms of Cancer Cachexia. Cancers (Basel) 2021; 13:cancers13163990. [PMID: 34439145 PMCID: PMC8391721 DOI: 10.3390/cancers13163990] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 08/05/2021] [Indexed: 12/04/2022] Open
Abstract
Simple Summary Cancer cachexia is a devastating wasting syndrome that occurs in many illnesses, with signs and symptoms including anorexia, weight loss, cognitive impairment and fatigue. The brain is capable of exerting overarching homeostatic control of whole-body metabolism and is increasingly being recognized as an important mediator of cancer cachexia. Given the increased recognition and discovery of neural mechanisms of cancer cachexia, we sought to provide an in-depth review and update of mechanisms by which the brain initiates and propagates cancer cachexia programs. Furthermore, recent work has identified new molecular mediators of cachexia that exert their effects through their direct interaction with the brain. Therefore, this review will summarize neural mechanisms of cachexia and discuss recently identified neural mediators of cancer cachexia. Abstract Nearly half of cancer patients suffer from cachexia, a metabolic syndrome characterized by progressive atrophy of fat and lean body mass. This state of excess catabolism decreases quality of life, ability to tolerate treatment and eventual survival, yet no effective therapies exist. Although the central nervous system (CNS) orchestrates several manifestations of cachexia, the precise mechanisms of neural dysfunction during cachexia are still being unveiled. Herein, we summarize the cellular and molecular mechanisms of CNS dysfunction during cancer cachexia with a focus on inflammatory, autonomic and neuroendocrine processes and end with a discussion of recently identified CNS mediators of cachexia, including GDF15, LCN2 and INSL3.
Collapse
Affiliation(s)
- Brennan Olson
- Medical Scientist Training Program, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA; (B.O.); (P.D.); (T.K.)
- Papé Family Pediatric Research Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA
| | - Parham Diba
- Medical Scientist Training Program, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA; (B.O.); (P.D.); (T.K.)
- Papé Family Pediatric Research Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA
| | - Tetiana Korzun
- Medical Scientist Training Program, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA; (B.O.); (P.D.); (T.K.)
- Papé Family Pediatric Research Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA
| | - Daniel L. Marks
- Papé Family Pediatric Research Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA
- Brenden-Colson Center for Pancreatic Care, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA
- Correspondence:
| |
Collapse
|
34
|
Drosophila Larval Models of Invasive Tumorigenesis for In Vivo Studies on Tumour/Peripheral Host Tissue Interactions during Cancer Cachexia. Int J Mol Sci 2021; 22:ijms22158317. [PMID: 34361081 PMCID: PMC8347517 DOI: 10.3390/ijms22158317] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/27/2021] [Accepted: 07/29/2021] [Indexed: 01/06/2023] Open
Abstract
Cancer cachexia is a common deleterious paraneoplastic syndrome that represents an area of unmet clinical need, partly due to its poorly understood aetiology and complex multifactorial nature. We have interrogated multiple genetically defined larval Drosophila models of tumourigenesis against key features of human cancer cachexia. Our results indicate that cachectic tissue wasting is dependent on the genetic characteristics of the tumour and demonstrate that host malnutrition or tumour burden are not sufficient to drive wasting. We show that JAK/STAT and TNF-α/Egr signalling are elevated in cachectic muscle and promote tissue wasting. Furthermore, we introduce a dual driver system that allows independent genetic manipulation of tumour and host skeletal muscle. Overall, we present a novel Drosophila larval paradigm to study tumour/host tissue crosstalk in vivo, which may contribute to future research in cancer cachexia and impact the design of therapeutic approaches for this pathology.
Collapse
|
35
|
Léopold P. Sizes, proportions and environment. C R Biol 2021; 344:165-175. [PMID: 34213854 DOI: 10.5802/crbiol.48] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 03/10/2021] [Indexed: 11/24/2022]
Abstract
The sizes of living organisms range over twenty orders of magnitude. Within the same species, the size of individuals also varies according to the environmental conditions to which they are subjected. From the studies conducted on organisms as diverse as the drosophila, the salamander or the mouse, laws and conserved mechanisms emerge that shed light on the fundamental aspects of growth, but also on more medical issues such as tissue regeneration, metabolic homeostasis and cancer.
Collapse
Affiliation(s)
- Pierre Léopold
- Institut Curie, PSL Research University, CNRS UMR3215, INSERM U934, UPMC Paris-Sorbonne, 26 Rue d'Ulm, 75005, Paris, France
| |
Collapse
|
36
|
|