1
|
Gomez A, Gonzalez S, Oke A, Luo J, Duong JB, Esquerra RM, Zimmerman T, Capponi S, Fung JC, Nystul TG. A High-Throughput Method for Quantifying Drosophila Fecundity. TOXICS 2024; 12:658. [PMID: 39330586 PMCID: PMC11436201 DOI: 10.3390/toxics12090658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 08/30/2024] [Accepted: 09/03/2024] [Indexed: 09/28/2024]
Abstract
The fruit fly, Drosophila melanogaster, is an experimentally tractable model system that has recently emerged as a powerful "new approach methodology" (NAM) for chemical safety testing. As oogenesis is well conserved at the molecular and cellular level, measurements of Drosophila fecundity can be useful for identifying chemicals that affect reproductive health across species. However, standard Drosophila fecundity assays have been difficult to perform in a high-throughput manner because experimental factors such as the physiological state of the flies and environmental cues must be carefully controlled to achieve consistent results. In addition, exposing flies to a large number of different experimental conditions (such as chemical additives in the diet) and manually counting the number of eggs laid to determine the impact on fecundity is time-consuming. We have overcome these challenges by combining a new multiwell fly culture strategy with a novel 3D-printed fly transfer device to rapidly and accurately transfer flies from one plate to another, the RoboCam, a low-cost, custom-built robotic camera to capture images of the wells automatically, and an image segmentation pipeline to automatically identify and quantify eggs. We show that this method is compatible with robust and consistent egg laying throughout the assay period and demonstrate that the automated pipeline for quantifying fecundity is very accurate (r2 = 0.98 for the correlation between the automated egg counts and the ground truth). In addition, we show that this method can be used to efficiently detect the effects on fecundity induced by dietary exposure to chemicals. Taken together, this strategy substantially increases the efficiency and reproducibility of high-throughput egg-laying assays that require exposing flies to multiple different media conditions.
Collapse
Affiliation(s)
- Andreana Gomez
- Department of Anatomy, University of California, San Francisco, CA 94143, USA
| | - Sergio Gonzalez
- Department of Biology, San Francisco State University, San Francisco, CA 94132, USA
- Center for Cellular Construction, San Francisco, CA 94158, USA
| | - Ashwini Oke
- OB/GYN Department, University of California, San Francisco, CA 94143, USA
- Center for Reproductive Sciences, University of California, San Francisco, CA 94143, USA
| | - Jiayu Luo
- Department of Biology, San Francisco State University, San Francisco, CA 94132, USA
- Center for Cellular Construction, San Francisco, CA 94158, USA
| | - Johnny B. Duong
- Center for Cellular Construction, San Francisco, CA 94158, USA
- Department of Chemistry and Biochemistry, San Francisco State University, San Francisco, CA 94132, USA
| | - Raymond M. Esquerra
- Center for Cellular Construction, San Francisco, CA 94158, USA
- Department of Chemistry and Biochemistry, San Francisco State University, San Francisco, CA 94132, USA
| | - Thomas Zimmerman
- Center for Cellular Construction, San Francisco, CA 94158, USA
- Department of Chemistry and Biochemistry, San Francisco State University, San Francisco, CA 94132, USA
- IBM Almaden Research Center, San Jose, CA 95120, USA
| | - Sara Capponi
- Center for Cellular Construction, San Francisco, CA 94158, USA
- IBM Almaden Research Center, San Jose, CA 95120, USA
| | - Jennifer C. Fung
- OB/GYN Department, University of California, San Francisco, CA 94143, USA
- Center for Reproductive Sciences, University of California, San Francisco, CA 94143, USA
- San Francisco EaRTH Center, University of California, San Francisco, CA 94143, USA
| | - Todd G. Nystul
- Department of Anatomy, University of California, San Francisco, CA 94143, USA
- OB/GYN Department, University of California, San Francisco, CA 94143, USA
- Center for Reproductive Sciences, University of California, San Francisco, CA 94143, USA
- San Francisco EaRTH Center, University of California, San Francisco, CA 94143, USA
| |
Collapse
|
2
|
Ruan ZR, Yu Z, Xing C, Chen EH. Inter-organ steroid hormone signaling promotes myoblast fusion via direct transcriptional regulation of a single key effector gene. Curr Biol 2024; 34:1438-1452.e6. [PMID: 38513654 PMCID: PMC11003854 DOI: 10.1016/j.cub.2024.02.056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 12/24/2023] [Accepted: 02/23/2024] [Indexed: 03/23/2024]
Abstract
Steroid hormones regulate tissue development and physiology by modulating the transcription of a broad spectrum of genes. In insects, the principal steroid hormones, ecdysteroids, trigger the expression of thousands of genes through a cascade of transcription factors (TFs) to coordinate developmental transitions such as larval molting and metamorphosis. However, whether ecdysteroid signaling can bypass transcriptional hierarchies to exert its function in individual developmental processes is unclear. Here, we report that a single non-TF effector gene mediates the transcriptional output of ecdysteroid signaling in Drosophila myoblast fusion, a critical step in muscle development and differentiation. Specifically, we show that the 20-hydroxyecdysone (commonly referred to as "ecdysone") secreted from an extraembryonic tissue, amnioserosa, acts on embryonic muscle cells to directly activate the expression of antisocial (ants), which encodes an essential scaffold protein enriched at the fusogenic synapse. Not only is ants transcription directly regulated by the heterodimeric ecdysone receptor complex composed of ecdysone receptor (EcR) and ultraspiracle (USP) via ecdysone-response elements but also more strikingly, expression of ants alone is sufficient to rescue the myoblast fusion defect in ecdysone signaling-deficient mutants. We further show that EcR/USP and a muscle-specific TF Twist synergistically activate ants expression in vitro and in vivo. Taken together, our study provides the first example of a steroid hormone directly activating the expression of a single key non-TF effector gene to regulate a developmental process via inter-organ signaling and provides a new paradigm for understanding steroid hormone signaling in other developmental and physiological processes.
Collapse
Affiliation(s)
- Zhi-Rong Ruan
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ze Yu
- McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Chao Xing
- McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Elizabeth H Chen
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
3
|
Delong LM, Ross AE. Open multi-organ communication device for easy interrogation of tissue slices. LAB ON A CHIP 2023; 23:3034-3049. [PMID: 37278087 PMCID: PMC10330603 DOI: 10.1039/d3lc00115f] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Here, we have developed an open multi-organ communication device that facilitates cellular and molecular communication between ex vivo organ slices. Measuring communication between organs is vital for understanding the mechanisms of health regulation yet remains difficult with current technology. Communication between organs along the gut-brain-immune axis is a key regulator of gut homeostasis. As a novel application of the device, we have used tissue slices from the Peyer's patch (PP) and mesenteric lymph node (MLN) due to their importance in gut immunity; however, any organ slices could be used here. The device was designed and fabricated using a combination of 3D printed molds for polydimethylsiloxane (PDMS) soft lithography, PDMS membranes, and track-etch porous membranes. To validate cellular and protein transfer between organs on-chip, we used fluorescence microscopy to quantitate movement of fluorescent proteins and cells from the PP to the MLN, replicating the initial response to immune stimuli in the gut. IFN-γ secretion during perfusion from a naïve vs. inflamed PP to a healthy MLN was quantitated to demonstrate soluble signaling molecules are moving on-chip. Finally, transient catecholamine release was measured during perfusion from PP to MLN using fast-scan cyclic voltammetry at carbon-fiber microelectrodes to demonstrate a novel application of the device for real-time sensing during communication. Overall, we show an open-well multi-organ device capable of facilitating transfer of soluble factors and cells with the added benefit of being available for external analysis techniques like electrochemical sensing which will advance abilities to probe communication in real-time across multiple organs ex vivo.
Collapse
Affiliation(s)
- Lauren M Delong
- Department of Chemistry, University of Cincinnati, 312 College Dr., 404 Crosley Tower, Cincinnati, OH 45221-0172, USA.
| | - Ashley E Ross
- Department of Chemistry, University of Cincinnati, 312 College Dr., 404 Crosley Tower, Cincinnati, OH 45221-0172, USA.
| |
Collapse
|
4
|
Xiong C, Zhou Y, Han Y, Yi J, Pang H, Zheng R, Zhou Y. IntiCom-DB: A Manually Curated Database of Inter-Tissue Communication Molecules and Their Communication Routes. BIOLOGY 2023; 12:833. [PMID: 37372118 DOI: 10.3390/biology12060833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 05/31/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023]
Abstract
Inter-tissue communication (ITC) is critical for maintaining the physiological functions of multiple tissues and is closely related to the onset and development of various complex diseases. Nevertheless, there is no well-organized data resource for known ITC molecules with explicit ITC routes from source tissues to target tissues. To address this issue, in this work, we manually reviewed nearly 190,000 publications and identified 1408 experimentally supported ITC entries in which the ITC molecules, their communication routes, and their functional annotations were included. To facilitate our work, these curated ITC entries were incorporated into a user-friendly database named IntiCom-DB. This database also enables visualization of the expression abundances of ITC proteins and their interaction partners. Finally, bioinformatics analyses on these data revealed common biological characteristics of the ITC molecules. For example, tissue specificity scores of ITC molecules at the protein level are often higher than those at the mRNA level in the target tissues. Moreover, the ITC molecules and their interaction partners are more abundant in both the source tissues and the target tissues. IntiCom-DB is freely available as an online database. As the first comprehensive database of ITC molecules with explicit ITC routes to the best of our knowledge, we hope that IntiCom-DB will benefit future ITC-related studies.
Collapse
Affiliation(s)
- Changxian Xiong
- Department of Biomedical Informatics, Center for Noncoding RNA Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| | - Yiran Zhou
- Department of Biomedical Informatics, Center for Noncoding RNA Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Yu Han
- Department of Biomedical Informatics, Center for Noncoding RNA Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| | - Jingkun Yi
- Department of Biomedical Informatics, Center for Noncoding RNA Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| | - Huai Pang
- Department of Biomedical Informatics, Center for Noncoding RNA Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| | - Ruimao Zheng
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Yuan Zhou
- Department of Biomedical Informatics, Center for Noncoding RNA Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| |
Collapse
|
5
|
Kumar T, Sethuraman R, Mitra S, Ravindran B, Narayanan M. MultiCens: Multilayer network centrality measures to uncover molecular mediators of tissue-tissue communication. PLoS Comput Biol 2023; 19:e1011022. [PMID: 37093889 PMCID: PMC10159362 DOI: 10.1371/journal.pcbi.1011022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 05/04/2023] [Accepted: 03/12/2023] [Indexed: 04/25/2023] Open
Abstract
With the evolution of multicellularity, communication among cells in different tissues and organs became pivotal to life. Molecular basis of such communication has long been studied, but genome-wide screens for genes and other biomolecules mediating tissue-tissue signaling are lacking. To systematically identify inter-tissue mediators, we present a novel computational approach MultiCens (Multilayer/Multi-tissue network Centrality measures). Unlike single-layer network methods, MultiCens can distinguish within- vs. across-layer connectivity to quantify the "influence" of any gene in a tissue on a query set of genes of interest in another tissue. MultiCens enjoys theoretical guarantees on convergence and decomposability, and performs well on synthetic benchmarks. On human multi-tissue datasets, MultiCens predicts known and novel genes linked to hormones. MultiCens further reveals shifts in gene network architecture among four brain regions in Alzheimer's disease. MultiCens-prioritized hypotheses from these two diverse applications, and potential future ones like "Multi-tissue-expanded Gene Ontology" analysis, can enable whole-body yet molecular-level systems investigations in humans.
Collapse
Affiliation(s)
- Tarun Kumar
- Department of Computer Science and Engineering, Indian Institute of Technology (IIT) Madras, Chennai, India
- The Centre for Integrative Biology and Systems medicinE (IBSE), IIT Madras, Chennai, India
- Robert Bosch Center for Data Science and Artificial Intelligence (RBCDSAI), IIT Madras, Chennai, India
| | | | - Sanga Mitra
- Department of Computer Science and Engineering, Indian Institute of Technology (IIT) Madras, Chennai, India
| | - Balaraman Ravindran
- Department of Computer Science and Engineering, Indian Institute of Technology (IIT) Madras, Chennai, India
- The Centre for Integrative Biology and Systems medicinE (IBSE), IIT Madras, Chennai, India
- Robert Bosch Center for Data Science and Artificial Intelligence (RBCDSAI), IIT Madras, Chennai, India
| | - Manikandan Narayanan
- Department of Computer Science and Engineering, Indian Institute of Technology (IIT) Madras, Chennai, India
- The Centre for Integrative Biology and Systems medicinE (IBSE), IIT Madras, Chennai, India
- Robert Bosch Center for Data Science and Artificial Intelligence (RBCDSAI), IIT Madras, Chennai, India
- Multiscale Digital Neuroanatomy (MDN), IIT Madras, Chennai, India
| |
Collapse
|
6
|
Gainullina A, Mogilenko DA, Huang LH, Todorov H, Narang V, Kim KW, Yng LS, Kent A, Jia B, Seddu K, Krchma K, Wu J, Crozat K, Tomasello E, Dress R, See P, Scott C, Gibbings S, Bajpai G, Desai JV, Maier B, This S, Wang P, Aguilar SV, Poupel L, Dussaud S, Zhou TA, Angeli V, Blander JM, Choi K, Dalod M, Dzhagalov I, Gautier EL, Jakubzick C, Lavine K, Lionakis MS, Paidassi H, Sieweke MH, Ginhoux F, Guilliams M, Benoist C, Merad M, Randolph GJ, Sergushichev A, Artyomov MN. Network analysis of large-scale ImmGen and Tabula Muris datasets highlights metabolic diversity of tissue mononuclear phagocytes. Cell Rep 2023; 42:112046. [PMID: 36708514 PMCID: PMC10372199 DOI: 10.1016/j.celrep.2023.112046] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 12/06/2022] [Accepted: 01/17/2023] [Indexed: 01/28/2023] Open
Abstract
The diversity of mononuclear phagocyte (MNP) subpopulations across tissues is one of the key physiological characteristics of the immune system. Here, we focus on understanding the metabolic variability of MNPs through metabolic network analysis applied to three large-scale transcriptional datasets: we introduce (1) an ImmGen MNP open-source dataset of 337 samples across 26 tissues; (2) a myeloid subset of ImmGen Phase I dataset (202 MNP samples); and (3) a myeloid mouse single-cell RNA sequencing (scRNA-seq) dataset (51,364 cells) assembled based on Tabula Muris Senis. To analyze such large-scale datasets, we develop a network-based computational approach, genes and metabolites (GAM) clustering, for unbiased identification of the key metabolic subnetworks based on transcriptional profiles. We define 9 metabolic subnetworks that encapsulate the metabolic differences within MNP from 38 different tissues. Obtained modules reveal that cholesterol synthesis appears particularly active within the migratory dendritic cells, while glutathione synthesis is essential for cysteinyl leukotriene production by peritoneal and lung macrophages.
Collapse
Affiliation(s)
- Anastasiia Gainullina
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA; Computer Technologies Department, ITMO University, St. Petersburg 197101, Russia; Laboratory of Bioinformatics and Molecular Genetics, Koltzov Institute of Developmental Biology of the Russian Academy of Sciences, Moscow 119334, Russia
| | - Denis A Mogilenko
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Medicine, Department of Pathology, Microbiology, and Immunology, Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Li-Hao Huang
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Helena Todorov
- Laboratory of Immunoregulation, Inflammation Research Centre, VIB Ghent University, 9052 Ghent, Belgium
| | - Vipin Narang
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A∗STAR), Singapore 138648, Singapore
| | - Ki-Wook Kim
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Lim Sheau Yng
- Immunology Translational Research Programme, Yong Loo Lin School of Medicine, Department of Microbiology and Immunology, National University of Singapore, Singapore 117545, Singapore; Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore 117545, Singapore
| | - Andrew Kent
- The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Joan and Sanford I. Weill Department of Medicine, Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
| | - Baosen Jia
- The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Joan and Sanford I. Weill Department of Medicine, Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
| | - Kumba Seddu
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Karen Krchma
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jun Wu
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Karine Crozat
- Aix Marseille University, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-Luminy, Turing Center for Living Systems, 13288 Marseille, France
| | - Elena Tomasello
- Aix Marseille University, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-Luminy, Turing Center for Living Systems, 13288 Marseille, France
| | - Regine Dress
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A∗STAR), Singapore 138648, Singapore
| | - Peter See
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A∗STAR), Singapore 138648, Singapore
| | - Charlotte Scott
- Laboratory of Immunoregulation, Inflammation Research Centre, VIB Ghent University, 9052 Ghent, Belgium
| | - Sophie Gibbings
- Department of Pediatrics, National Jewish Health, Denver, CO 80206, USA
| | - Geetika Bajpai
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Jigar V Desai
- Fungal Pathogenesis Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Barbara Maier
- Immunology Institute and Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sébastien This
- Centre International de Recherche en Infectiologie (CIRI), University Lyon, Inserm, U1111, Université Claude Bernard Lyon ,1, CNRS, UMR5308, ENS de Lyon, 69007 Lyon, France
| | - Peter Wang
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Stephanie Vargas Aguilar
- Aix Marseille University, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-Luminy, Turing Center for Living Systems, 13288 Marseille, France; Center for Regenerative Therapies (CRTD), TU Dresden, 01307 Dresden, Germany; Max-Delbrück-Centrum für Molekulare Medizin in der Helmholtzgemeinschaft (MDC), 13125 Berlin, Germany
| | - Lucie Poupel
- INSERM UMR-S 1166, Sorbonne Université, Hôpital de la Pitié-Salpêtrière, 75013 Paris, France
| | - Sébastien Dussaud
- INSERM UMR-S 1166, Sorbonne Université, Hôpital de la Pitié-Salpêtrière, 75013 Paris, France
| | - Tyng-An Zhou
- Institute of Microbiology and Immunology, National Yang-Ming University, Taipei 112, Taiwan
| | - Veronique Angeli
- Immunology Translational Research Programme, Yong Loo Lin School of Medicine, Department of Microbiology and Immunology, National University of Singapore, Singapore 117545, Singapore; Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore 117545, Singapore
| | - J Magarian Blander
- The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Joan and Sanford I. Weill Department of Medicine, Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
| | - Kyunghee Choi
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Marc Dalod
- Aix Marseille University, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-Luminy, Turing Center for Living Systems, 13288 Marseille, France
| | - Ivan Dzhagalov
- Institute of Microbiology and Immunology, National Yang-Ming University, Taipei 112, Taiwan
| | - Emmanuel L Gautier
- INSERM UMR-S 1166, Sorbonne Université, Hôpital de la Pitié-Salpêtrière, 75013 Paris, France
| | - Claudia Jakubzick
- Department of Pediatrics, National Jewish Health, Denver, CO 80206, USA
| | - Kory Lavine
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Michail S Lionakis
- Fungal Pathogenesis Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Helena Paidassi
- Centre International de Recherche en Infectiologie (CIRI), University Lyon, Inserm, U1111, Université Claude Bernard Lyon ,1, CNRS, UMR5308, ENS de Lyon, 69007 Lyon, France
| | - Michael H Sieweke
- Aix Marseille University, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-Luminy, Turing Center for Living Systems, 13288 Marseille, France; Center for Regenerative Therapies (CRTD), TU Dresden, 01307 Dresden, Germany; Max-Delbrück-Centrum für Molekulare Medizin in der Helmholtzgemeinschaft (MDC), 13125 Berlin, Germany
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A∗STAR), Singapore 138648, Singapore
| | - Martin Guilliams
- Laboratory of Immunoregulation, Inflammation Research Centre, VIB Ghent University, 9052 Ghent, Belgium
| | | | - Miriam Merad
- Immunology Institute and Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Gwendalyn J Randolph
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Alexey Sergushichev
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA; Computer Technologies Department, ITMO University, St. Petersburg 197101, Russia.
| | - Maxim N Artyomov
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
7
|
Mehta RI, Mehta RI. The Vascular-Immune Hypothesis of Alzheimer's Disease. Biomedicines 2023; 11:408. [PMID: 36830944 PMCID: PMC9953491 DOI: 10.3390/biomedicines11020408] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 01/26/2023] [Accepted: 01/27/2023] [Indexed: 01/31/2023] Open
Abstract
Alzheimer's disease (AD) is a devastating and irreversible neurodegenerative disorder with unknown etiology. While its cause is unclear, a number of theories have been proposed to explain the pathogenesis of AD. In large part, these have centered around potential causes for intracerebral accumulation of beta-amyloid (βA) and tau aggregates. Yet, persons with AD dementia often exhibit autopsy evidence of mixed brain pathologies including a myriad of vascular changes, vascular brain injuries, complex brain inflammation, and mixed protein inclusions in addition to hallmark neuropathologic lesions of AD, namely insoluble βA plaques and neurofibrillary tangles (NFTs). Epidemiological data demonstrate that overlapping lesions diminish the βA plaque and NFT threshold necessary to precipitate clinical dementia. Moreover, a subset of persons who exhibit AD pathology remain resilient to disease while other persons with clinically-defined AD dementia do not exhibit AD-defining neuropathologic lesions. It is increasingly recognized that AD is a pathologically heterogeneous and biologically multifactorial disease with uncharacterized biologic phenomena involved in its genesis and progression. Here, we review the literature with regard to neuropathologic criteria and incipient AD changes, and discuss converging concepts regarding vascular and immune factors in AD.
Collapse
Affiliation(s)
- Rashi I. Mehta
- Department of Neuroradiology, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506, USA
| | - Rupal I. Mehta
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL 60612, USA
- Department of Pathology, Rush University Medical Center, Chicago, IL 60612, USA
| |
Collapse
|
8
|
Berthou F, Sobolewski C, Abegg D, Fournier M, Maeder C, Dolicka D, Correia de Sousa M, Adibekian A, Foti M. Hepatic PTEN Signaling Regulates Systemic Metabolic Homeostasis through Hepatokines-Mediated Liver-to-Peripheral Organs Crosstalk. Int J Mol Sci 2022; 23:ijms23073959. [PMID: 35409319 PMCID: PMC8999584 DOI: 10.3390/ijms23073959] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 03/25/2022] [Accepted: 03/31/2022] [Indexed: 11/16/2022] Open
Abstract
Liver-derived circulating factors deeply affect the metabolism of distal organs. Herein, we took advantage of the hepatocyte-specific PTEN knockout mice (LPTENKO), a model of hepatic steatosis associated with increased muscle insulin sensitivity and decreased adiposity, to identify potential secreted hepatic factors improving metabolic homeostasis. Our results indicated that protein factors, rather than specific metabolites, released by PTEN-deficient hepatocytes trigger an improved muscle insulin sensitivity and a decreased adiposity in LPTENKO. In this regard, a proteomic analysis of conditioned media from PTEN-deficient primary hepatocytes identified seven hepatokines whose expression/secretion was deregulated. Distinct expression patterns of these hepatokines were observed in hepatic tissues from human/mouse with NAFLD. The expression of specific factors was regulated by the PTEN/PI3K, PPAR or AMPK signaling pathways and/or modulated by classical antidiabetic drugs. Finally, loss-of-function studies identified FGF21 and the triad AHSG, ANGPTL4 and LECT2 as key regulators of insulin sensitivity in muscle cells and in adipocytes biogenesis, respectively. These data indicate that hepatic PTEN deficiency and steatosis alter the expression/secretion of hepatokines regulating insulin sensitivity in muscles and the lipid metabolism in adipose tissue. These hepatokines could represent potential therapeutic targets to treat obesity and insulin resistance.
Collapse
Affiliation(s)
- Flavien Berthou
- Department of Cellular Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1206 Geneva, Switzerland; (F.B.); (C.S.); (M.F.); (C.M.); (D.D.); (M.C.d.S.)
| | - Cyril Sobolewski
- Department of Cellular Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1206 Geneva, Switzerland; (F.B.); (C.S.); (M.F.); (C.M.); (D.D.); (M.C.d.S.)
| | - Daniel Abegg
- Department of Chemistry, The Scripps Research Institute, Jupiter, FL 33458, USA; (D.A.); (A.A.)
| | - Margot Fournier
- Department of Cellular Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1206 Geneva, Switzerland; (F.B.); (C.S.); (M.F.); (C.M.); (D.D.); (M.C.d.S.)
| | - Christine Maeder
- Department of Cellular Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1206 Geneva, Switzerland; (F.B.); (C.S.); (M.F.); (C.M.); (D.D.); (M.C.d.S.)
| | - Dobrochna Dolicka
- Department of Cellular Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1206 Geneva, Switzerland; (F.B.); (C.S.); (M.F.); (C.M.); (D.D.); (M.C.d.S.)
| | - Marta Correia de Sousa
- Department of Cellular Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1206 Geneva, Switzerland; (F.B.); (C.S.); (M.F.); (C.M.); (D.D.); (M.C.d.S.)
| | - Alexander Adibekian
- Department of Chemistry, The Scripps Research Institute, Jupiter, FL 33458, USA; (D.A.); (A.A.)
| | - Michelangelo Foti
- Department of Cellular Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1206 Geneva, Switzerland; (F.B.); (C.S.); (M.F.); (C.M.); (D.D.); (M.C.d.S.)
- Diabetes Center, Faculty of Medicine, University of Geneva, 1206 Geneva, Switzerland
- Correspondence: ; Tel.: +41-(22)-379-52-04
| |
Collapse
|
9
|
Herrlich A. Interorgan crosstalk mechanisms in disease: the case of acute kidney injury-induced remote lung injury. FEBS Lett 2021; 596:620-637. [PMID: 34932216 DOI: 10.1002/1873-3468.14262] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 12/08/2021] [Accepted: 12/09/2021] [Indexed: 11/07/2022]
Abstract
Homeostasis and health of multicellular organisms with multiple organs depends on interorgan communication. Tissue injury in one organ disturbs this homeostasis and can lead to disease in multiple organs, or multiorgan failure. Many routes of interorgan crosstalk during homeostasis are relatively well known, but interorgan crosstalk in disease still lacks understanding. In particular, how tissue injury in one organ can drive injury at remote sites and trigger multiorgan failure with high mortality is poorly understood. As examples, acute kidney injury can trigger acute lung injury and cardiovascular dysfunction; pneumonia, sepsis or liver failure conversely can cause kidney failure; lung transplantation very frequently triggers acute kidney injury. Mechanistically, interorgan crosstalk after tissue injury could involve soluble mediators and their target receptors, cellular mediators, in particular immune cells, as well as newly identified neuro-immune connections. In this review, I will focus the discussion of deleterious interorgan crosstalk and its mechanistic concepts on one example, acute kidney injury-induced remote lung injury.
Collapse
Affiliation(s)
- Andreas Herrlich
- Division of Nephrology, Department of Medicine, Washington University in St. Louis, MO, USA
| |
Collapse
|
10
|
Pasquier C, Robichon A. Temporal and sequential order of nonoverlapping gene networks unraveled in mated female Drosophila. Life Sci Alliance 2021; 5:5/2/e202101119. [PMID: 34844981 PMCID: PMC8645335 DOI: 10.26508/lsa.202101119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 11/11/2021] [Accepted: 11/12/2021] [Indexed: 12/13/2022] Open
Abstract
Mating triggers successive waves of temporal transcriptomic changes within independent gene networks in female Drosophila, suggesting a recruitment of interconnected modules that vanish in late life. In this study, we reanalyzed available datasets of gene expression changes in female Drosophila head induced by mating. Mated females present metabolic phenotypic changes and display behavioral characteristics that are not observed in virgin females, such as repulsion to male sexual aggressiveness, fidelity to food spots selected for oviposition, and restriction to the colonization of new niches. We characterize gene networks that play a role in female brain plasticity after mating using AMINE, a novel algorithm to find dysregulated modules of interacting genes. The uncovered networks of altered genes revealed a strong specificity for each successive period of life span after mating in the female head, with little conservation between them. This finding highlights a temporal order of recruitment of waves of interconnected genes which are apparently transiently modified: the first wave disappears before the emergence of the second wave in a reversible manner and ends with few consolidated gene expression changes at day 20. This analysis might document an extended field of a programmatic control of female phenotypic traits by male seminal fluid.
Collapse
|
11
|
Paul D, Komarova NL. Multi-scale network targeting: A holistic systems-biology approach to cancer treatment. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2021; 165:72-79. [PMID: 34428429 DOI: 10.1016/j.pbiomolbio.2021.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 08/05/2021] [Accepted: 08/10/2021] [Indexed: 11/15/2022]
Abstract
The vulnerabilities of cancer at the cellular and, recently, with the introduction of immunotherapy, at the tissue level, have been exploited with variable success. Evaluating the cancer system vulnerabilities at the organismic level through analysis of network topology and network dynamics can potentially predict novel anti-cancer drug targets directed at the macroscopic cancer networks. Theoretical work analyzing the properties and the vulnerabilities of the multi-scale network of cancer needs to go hand-in-hand with experimental research that uncovers the biological nature of the relevant networks and reveals new targetable vulnerabilities. It is our hope that attacking cancer on different spatial scales, in a concerted integrated approach, may present opportunities for novel ways to prevent treatment resistance.
Collapse
Affiliation(s)
- Doru Paul
- Medical Oncology, Weill Cornell Medicine, 1305 York Avenue 12th Floor, New York, NY, 10021, USA.
| | - Natalia L Komarova
- Department of Mathematics, University of California Irvine, Irvine, CA, 92697, USA.
| |
Collapse
|
12
|
Bodine SC, Brooks HL, Bunnett NW, Coller HA, Frey MR, Joe B, Kleyman TR, Lindsey ML, Marette A, Morty RE, Ramírez JM, Thomsen MB, Yosten GLC. An American Physiological Society cross-journal Call for Papers on "Inter-Organ Communication in Homeostasis and Disease". Am J Physiol Lung Cell Mol Physiol 2021; 321:L42-L49. [PMID: 34010064 PMCID: PMC8321848 DOI: 10.1152/ajplung.00209.2021] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 05/13/2021] [Accepted: 05/15/2021] [Indexed: 12/17/2022] Open
Affiliation(s)
- Sue C Bodine
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Heddwen L Brooks
- Department of Physiology, University of Arizona College of Medicine, Tucson, Arizona
| | - Nigel W Bunnett
- Department of Molecular Pathobiology, New York University, New York, New York
| | - Hilary A Coller
- Molecular Biology Interdepartmental Program, University of California, Los Angeles, California
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, California
- Department of Biological Chemistry, University of California, Los Angeles, California
| | - Mark R Frey
- The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California
- Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, California
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Bina Joe
- Department of Physiology and Pharmacology, College of Medicine and Life Sciences, University of Toledo, Toledo, Ohio
- Center for Hypertension and Personalized Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, Ohio
| | - Thomas R Kleyman
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Merry L Lindsey
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, Nebraska
- Research Service, VA Nebraska-Western Iowa Health Care System, Omaha, Nebraska
| | - André Marette
- Department of Medicine, Faculty of Medicine, Cardiology Axis of the Québec Heart and Lung Institute, Hôpital Laval, Laval University, Quebec City, Québec, Canada
- Institute of Nutrition and Functional Foods, Laval University, Quebec City, Québec, Canada
| | - Rory E Morty
- Department of Translational Pulmonology and the Translational Lung Research Center Heidelberg, University Hospital Heidelberg, member of the German Center for Lung Research (DZL), Heidelberg, Germany
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, Justus Liebig University Giessen, member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Jan-Marino Ramírez
- Department of Neurological Surgery, University of Washington Medical Center, Seattle, Washington
- Center on Human Development and Disability, University of Washington, Seattle, Washington
- Center for Integrative Brain Research at the Seattle Children's Research Institute, University of Washington, Seattle, Washington
| | - Morten B Thomsen
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Gina L C Yosten
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri
- Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, St. Louis, Missouri
| |
Collapse
|
13
|
Solagna F, Tezze C, Lindenmeyer MT, Lu S, Wu G, Liu S, Zhao Y, Mitchell R, Meyer C, Omairi S, Kilic T, Paolini A, Ritvos O, Pasternack A, Matsakas A, Kylies D, zur Wiesch JS, Turner JE, Wanner N, Nair V, Eichinger F, Menon R, Martin IV, Klinkhammer BM, Hoxha E, Cohen CD, Tharaux PL, Boor P, Ostendorf T, Kretzler M, Sandri M, Kretz O, Puelles VG, Patel K, Huber TB. Pro-cachectic factors link experimental and human chronic kidney disease to skeletal muscle wasting programs. J Clin Invest 2021; 131:135821. [PMID: 34060483 PMCID: PMC8159690 DOI: 10.1172/jci135821] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 04/21/2021] [Indexed: 02/06/2023] Open
Abstract
Skeletal muscle wasting is commonly associated with chronic kidney disease (CKD), resulting in increased morbidity and mortality. However, the link between kidney and muscle function remains poorly understood. Here, we took a complementary interorgan approach to investigate skeletal muscle wasting in CKD. We identified increased production and elevated blood levels of soluble pro-cachectic factors, including activin A, directly linking experimental and human CKD to skeletal muscle wasting programs. Single-cell sequencing data identified the expression of activin A in specific kidney cell populations of fibroblasts and cells of the juxtaglomerular apparatus. We propose that persistent and increased kidney production of pro-cachectic factors, combined with a lack of kidney clearance, facilitates a vicious kidney/muscle signaling cycle, leading to exacerbated blood accumulation and, thereby, skeletal muscle wasting. Systemic pharmacological blockade of activin A using soluble activin receptor type IIB ligand trap as well as muscle-specific adeno-associated virus-mediated downregulation of its receptor ACVR2A/B prevented muscle wasting in different mouse models of experimental CKD, suggesting that activin A is a key factor in CKD-induced cachexia. In summary, we uncovered a crosstalk between kidney and muscle and propose modulation of activin signaling as a potential therapeutic strategy for skeletal muscle wasting in CKD.
Collapse
Affiliation(s)
- Francesca Solagna
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Caterina Tezze
- Veneto Institute of Molecular Medicine, Padua, Italy
- Department of Biomedical Sciences, University of Padova, Padua, Italy
| | - Maja T. Lindenmeyer
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Shun Lu
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Guochao Wu
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Shuya Liu
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Yu Zhao
- Institute of Medical Systems Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Robert Mitchell
- School of Biological Sciences, University of Reading, Reading, United Kingdom
| | - Charlotte Meyer
- Renal Division, Faculty of Medicine, Medical Centre, University of Freiburg, Freiburg, Germany
| | - Saleh Omairi
- College of Medicine, University of Wasit, Kut, Iraq
| | - Temel Kilic
- Renal Division, Faculty of Medicine, Medical Centre, University of Freiburg, Freiburg, Germany
| | - Andrea Paolini
- School of Biological Sciences, University of Reading, Reading, United Kingdom
| | - Olli Ritvos
- Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Arja Pasternack
- Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Antonios Matsakas
- Molecular Physiology Laboratory, Centre for Atherothrombosis and Metabolic Disease, Hull York Medical School, Hull, United Kingdom
| | - Dominik Kylies
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Jan-Eric Turner
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nicola Wanner
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Viji Nair
- Michigan Medicine, Ann Arbor, Michigan, USA
| | | | | | - Ina V. Martin
- Department of Nephrology and Clinical Immunology and
| | | | - Elion Hoxha
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Clemens D. Cohen
- Nephrological Center, Medical Clinic and Polyclinic IV, University of Munich, Munich, Germany
| | - Pierre-Louis Tharaux
- Paris Centre de Recherche Cardiovasculaire, INSERM, Université de Paris, Paris, France
| | - Peter Boor
- Department of Nephrology and Clinical Immunology and
- Institute of Pathology, University Hospital RWTH Aachen, Aachen, Germany
| | | | | | - Marco Sandri
- Veneto Institute of Molecular Medicine, Padua, Italy
- Department of Biomedical Sciences, University of Padova, Padua, Italy
| | - Oliver Kretz
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Victor G. Puelles
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Anatomy and Developmental Biology, Monash University, Melbourne, Australia
| | - Ketan Patel
- School of Biological Sciences, University of Reading, Reading, United Kingdom
- Freiburg Institute for Advanced Studies and Center for Biological System Analysis, University of Freiburg, Freiburg, Germany
| | - Tobias B. Huber
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Freiburg Institute for Advanced Studies and Center for Biological System Analysis, University of Freiburg, Freiburg, Germany
| |
Collapse
|
14
|
Zandawala M, Nguyen T, Balanyà Segura M, Johard HAD, Amcoff M, Wegener C, Paluzzi JP, Nässel DR. A neuroendocrine pathway modulating osmotic stress in Drosophila. PLoS Genet 2021; 17:e1009425. [PMID: 33684132 PMCID: PMC7971876 DOI: 10.1371/journal.pgen.1009425] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 03/18/2021] [Accepted: 02/15/2021] [Indexed: 12/19/2022] Open
Abstract
Environmental factors challenge the physiological homeostasis in animals, thereby evoking stress responses. Various mechanisms have evolved to counter stress at the organism level, including regulation by neuropeptides. In recent years, much progress has been made on the mechanisms and neuropeptides that regulate responses to metabolic/nutritional stress, as well as those involved in countering osmotic and ionic stresses. Here, we identified a peptidergic pathway that links these types of regulatory functions. We uncover the neuropeptide Corazonin (Crz), previously implicated in responses to metabolic stress, as a neuroendocrine factor that inhibits the release of a diuretic hormone, CAPA, and thereby modulates the tolerance to osmotic and ionic stress. Both knockdown of Crz and acute injections of Crz peptide impact desiccation tolerance and recovery from chill-coma. Mapping of the Crz receptor (CrzR) expression identified three pairs of Capa-expressing neurons (Va neurons) in the ventral nerve cord that mediate these effects of Crz. We show that Crz acts to restore water/ion homeostasis by inhibiting release of CAPA neuropeptides via inhibition of cAMP production in Va neurons. Knockdown of CrzR in Va neurons affects CAPA signaling, and consequently increases tolerance for desiccation, ionic stress and starvation, but delays chill-coma recovery. Optogenetic activation of Va neurons stimulates excretion and simultaneous activation of Crz and CAPA-expressing neurons reduces this response, supporting the inhibitory action of Crz. Thus, Crz inhibits Va neurons to maintain osmotic and ionic homeostasis, which in turn affects stress tolerance. Earlier work demonstrated that systemic Crz signaling restores nutrient levels by promoting food search and feeding. Here we additionally propose that Crz signaling also ensures osmotic homeostasis by inhibiting release of CAPA neuropeptides and suppressing diuresis. Thus, Crz ameliorates stress-associated physiology through systemic modulation of both peptidergic neurosecretory cells and the fat body in Drosophila.
Collapse
Affiliation(s)
- Meet Zandawala
- Department of Zoology, Stockholm University, Stockholm, Sweden
| | - Thomas Nguyen
- Department of Biology, York University, Toronto, Ontario, Canada
| | - Marta Balanyà Segura
- Neurobiology and Genetics, Würzburg Insect Research (WIR), Theodor-Boveri-Institute, Biocenter, University of Würzburg, Germany
| | | | - Mirjam Amcoff
- Department of Zoology, Stockholm University, Stockholm, Sweden
| | - Christian Wegener
- Neurobiology and Genetics, Würzburg Insect Research (WIR), Theodor-Boveri-Institute, Biocenter, University of Würzburg, Germany
| | | | - Dick R Nässel
- Department of Zoology, Stockholm University, Stockholm, Sweden
| |
Collapse
|
15
|
Girousse A, Mathieu M, Sastourné-Arrey Q, Monferran S, Casteilla L, Sengenès C. Endogenous Mobilization of Mesenchymal Stromal Cells: A Pathway for Interorgan Communication? Front Cell Dev Biol 2021; 8:598520. [PMID: 33490065 PMCID: PMC7820193 DOI: 10.3389/fcell.2020.598520] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 11/18/2020] [Indexed: 12/16/2022] Open
Abstract
To coordinate specialized organs, inter-tissue communication appeared during evolution. Consequently, individual organs communicate their states via a vast interorgan communication network (ICN) made up of peptides, proteins, and metabolites that act between organs to coordinate cellular processes under homeostasis and stress. However, the nature of the interorgan signaling could be even more complex and involve mobilization mechanisms of unconventional cells that are still poorly described. Mesenchymal stem/stromal cells (MSCs) virtually reside in all tissues, though the biggest reservoir discovered so far is adipose tissue where they are named adipose stromal cells (ASCs). MSCs are thought to participate in tissue maintenance and repair since the administration of exogenous MSCs is well known to exert beneficial effects under several pathological conditions. However, the role of endogenous MSCs is barely understood. Though largely debated, the presence of circulating endogenous MSCs has been reported in multiple pathophysiological conditions, but the significance of such cell circulation is not known and therapeutically untapped. In this review, we discuss current knowledge on the circulation of native MSCs, and we highlight recent findings describing MSCs as putative key components of the ICN.
Collapse
Affiliation(s)
- Amandine Girousse
- Stromalab, Université de Toulouse, CNRS ERL5311, EFS, INP-ENVT, INSERM U1031, Université Paul Sabatier, Toulouse, France
| | - Maxime Mathieu
- Stromalab, Université de Toulouse, CNRS ERL5311, EFS, INP-ENVT, INSERM U1031, Université Paul Sabatier, Toulouse, France
| | - Quentin Sastourné-Arrey
- Sprott Center for Stem Cell Research, Ottawa Hospital Research Institute, Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Sylvie Monferran
- Stromalab, Université de Toulouse, CNRS ERL5311, EFS, INP-ENVT, INSERM U1031, Université Paul Sabatier, Toulouse, France
| | - Louis Casteilla
- Stromalab, Université de Toulouse, CNRS ERL5311, EFS, INP-ENVT, INSERM U1031, Université Paul Sabatier, Toulouse, France
| | - Coralie Sengenès
- Stromalab, Université de Toulouse, CNRS ERL5311, EFS, INP-ENVT, INSERM U1031, Université Paul Sabatier, Toulouse, France
| |
Collapse
|
16
|
Upadhyay A, Peterson AJ, Kim MJ, O'Connor MB. Muscle-derived Myoglianin regulates Drosophila imaginal disc growth. eLife 2020; 9:e51710. [PMID: 32633716 PMCID: PMC7371420 DOI: 10.7554/elife.51710] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Accepted: 07/04/2020] [Indexed: 01/05/2023] Open
Abstract
Organ growth and size are finely tuned by intrinsic and extrinsic signaling molecules. In Drosophila, the BMP family member Dpp is produced in a limited set of imaginal disc cells and functions as a classic morphogen to regulate pattern and growth by diffusing throughout imaginal discs. However, the role of TGFβ/Activin-like ligands in disc growth control remains ill-defined. Here, we demonstrate that Myoglianin (Myo), an Activin family member, and a close homolog of mammalian Myostatin (Mstn), is a muscle-derived extrinsic factor that uses canonical dSmad2-mediated signaling to regulate wing size. We propose that Myo is a myokine that helps mediate an allometric relationship between muscles and their associated appendages.
Collapse
Affiliation(s)
- Ambuj Upadhyay
- Department of Genetics, Cell Biology and Development University of MinnesotaMinneapolisUnited States
| | - Aidan J Peterson
- Department of Genetics, Cell Biology and Development University of MinnesotaMinneapolisUnited States
| | - Myung-Jun Kim
- Department of Genetics, Cell Biology and Development University of MinnesotaMinneapolisUnited States
| | - Michael B O'Connor
- Department of Genetics, Cell Biology and Development University of MinnesotaMinneapolisUnited States
| |
Collapse
|
17
|
Sandhof CA, Hoppe SO, Druffel-Augustin S, Gallrein C, Kirstein J, Voisine C, Nussbaum-Krammer C. Reducing INS-IGF1 signaling protects against non-cell autonomous vesicle rupture caused by SNCA spreading. Autophagy 2020; 16:878-899. [PMID: 31354022 PMCID: PMC7144869 DOI: 10.1080/15548627.2019.1643657] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 07/02/2019] [Accepted: 07/11/2019] [Indexed: 01/28/2023] Open
Abstract
Aging is associated with a gradual decline of cellular proteostasis, giving rise to devastating protein misfolding diseases, such as Alzheimer disease (AD) or Parkinson disease (PD). These diseases often exhibit a complex pathology involving non-cell autonomous proteotoxic effects, which are still poorly understood. Using Caenorhabditis elegans we investigated how local protein misfolding is affecting neighboring cells and tissues showing that misfolded PD-associated SNCA/α-synuclein is accumulating in highly dynamic endo-lysosomal vesicles. Irrespective of whether being expressed in muscle cells or dopaminergic neurons, accumulated proteins were transmitted into the hypodermis with increasing age, indicating that epithelial cells might play a role in remote degradation when the local endo-lysosomal degradation capacity is overloaded. Cell biological and genetic approaches revealed that inter-tissue dissemination of SNCA was regulated by endo- and exocytosis (neuron/muscle to hypodermis) and basement membrane remodeling (muscle to hypodermis). Transferred SNCA conformers were, however, inefficiently cleared and induced endo-lysosomal membrane permeabilization. Remarkably, reducing INS (insulin)-IGF1 (insulin-like growth factor 1) signaling provided protection by maintaining endo-lysosomal integrity. This study suggests that the degradation of lysosomal substrates is coordinated across different tissues in metazoan organisms. Because the chronic dissemination of poorly degradable disease proteins into neighboring tissues exerts a non-cell autonomous toxicity, this implies that restoring endo-lysosomal function not only in cells with pathological inclusions, but also in apparently unaffected cell types might help to halt disease progression.Abbreviations: AD: Alzheimer disease; BM: basement membrane; BWM: body wall muscle; CEP: cephalic sensilla; CLEM: correlative light and electron microscopy; CTNS-1: cystinosin (lysosomal protein) homolog; DA: dopaminergic; DAF-2: abnormal dauer formation; ECM: extracellular matrix; FLIM: fluorescence lifetime imaging microscopy; fps: frames per second; GFP: green fluorescent protein; HPF: high pressure freezing; IGF1: insulin-like growth factor 1; INS: insulin; KD: knockdown; LMP: lysosomal membrane permeabilization; MVB: multivesicular body; NOC: nocodazole; PD: Parkinson disease; RFP: red fluorescent protein; RNAi: RNA interference; sfGFP: superfolder GFP; SNCA: synuclein alpha; TEM: transmission electron microscopy; TNTs: tunneling nanotubes; TCSPC: time correlated single photon counting; YFP: yellow fluorescent protein.
Collapse
Affiliation(s)
- Carl Alexander Sandhof
- Center for Molecular Biology of Heidelberg University (ZMBH) and German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Simon Oliver Hoppe
- Center for Molecular Biology of Heidelberg University (ZMBH) and German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Silke Druffel-Augustin
- Center for Molecular Biology of Heidelberg University (ZMBH) and German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Christian Gallrein
- Department of Molecular Physiology and Cell Biology, Leibniz-Institute for Molecular Pharmacology (FMP) im Forschungsverbund Berlin e.V, Berlin, Germany
| | - Janine Kirstein
- Department of Molecular Physiology and Cell Biology, Leibniz-Institute for Molecular Pharmacology (FMP) im Forschungsverbund Berlin e.V, Berlin, Germany
| | - Cindy Voisine
- Department of Biology, Northeastern Illinois University, Chicago, IL, USA
| | - Carmen Nussbaum-Krammer
- Center for Molecular Biology of Heidelberg University (ZMBH) and German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Heidelberg, Germany
| |
Collapse
|
18
|
The Drosophila Post-mating Response: Gene Expression and Behavioral Changes Reveal Perdurance and Variation in Cross-Tissue Interactions. G3-GENES GENOMES GENETICS 2020; 10:967-983. [PMID: 31907222 PMCID: PMC7056969 DOI: 10.1534/g3.119.400963] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Examining cross-tissue interactions is important for understanding physiology and homeostasis. In animals, the female gonad produces signaling molecules that act distally. We examine gene expression in Drosophila melanogaster female head tissues in 1) virgins without a germline compared to virgins with a germline, 2) post-mated females with and without a germline compared to virgins, and 3) post-mated females mated to males with and without a germline compared to virgins. In virgins, the absence of a female germline results in expression changes in genes with known roles in nutrient homeostasis. At one- and three-day(s) post-mating, genes that change expression are enriched with those that function in metabolic pathways, in all conditions. We systematically examine female post-mating impacts on sleep, food preference and re-mating, in the strains and time points used for gene expression analyses and compare to published studies. We show that post-mating, gene expression changes vary by strain, prompting us to examine variation in female re-mating. We perform a genome-wide association study that identifies several DNA polymorphisms, including four in/near Wnt signaling pathway genes. Together, these data reveal how gene expression and behavior in females are influenced by cross-tissue interactions, by examining the impact of mating, fertility, and genotype.
Collapse
|
19
|
Kozawa S, Sagawa F, Endo S, De Almeida GM, Mitsuishi Y, Sato TN. Predicting Human Clinical Outcomes Using Mouse Multi-Organ Transcriptome. iScience 2020; 23:100791. [PMID: 31928967 PMCID: PMC7033637 DOI: 10.1016/j.isci.2019.100791] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 10/17/2019] [Accepted: 12/16/2019] [Indexed: 12/27/2022] Open
Abstract
Approximately 90% of pre-clinically validated drugs fail in clinical trials owing to unanticipated clinical outcomes, costing over several hundred million US dollars per drug. Despite such critical importance, translating pre-clinical data to clinical outcomes remain a major challenge. Herein, we designed a modality-independent and unbiased approach to predict clinical outcomes of drugs. The approach exploits their multi-organ transcriptome patterns induced in mice and a unique mouse-transcriptome database "humanized" by machine learning algorithms and human clinical outcome datasets. The cross-validation with small-molecule, antibody, and peptide drugs shows effective and efficient identification of the previously known outcomes of 5,519 adverse events and 11,312 therapeutic indications. In addition, the approach is adaptable to deducing potential molecular mechanisms underlying these outcomes. Furthermore, the approach identifies previously unsuspected repositioning targets. These results, together with the fact that it requires no prior structural or mechanistic information of drugs, illustrate its versatile applications to drug development process.
Collapse
Affiliation(s)
- Satoshi Kozawa
- Karydo TherapeutiX, Inc., Kyoto, Japan; ERATO Sato Live Bio-Forecasting Project, Kyoto, Japan; The Thomas N. Sato BioMEC-X Laboratories, Advanced Telecommunications Research Institute International, 2-2-2 Hikaridai, Seika-cho, Soraku-gun, Kyoto 619-0288, Japan
| | - Fumihiko Sagawa
- Karydo TherapeutiX, Inc., Kyoto, Japan; ERATO Sato Live Bio-Forecasting Project, Kyoto, Japan; The Thomas N. Sato BioMEC-X Laboratories, Advanced Telecommunications Research Institute International, 2-2-2 Hikaridai, Seika-cho, Soraku-gun, Kyoto 619-0288, Japan
| | - Satsuki Endo
- Karydo TherapeutiX, Inc., Kyoto, Japan; ERATO Sato Live Bio-Forecasting Project, Kyoto, Japan; The Thomas N. Sato BioMEC-X Laboratories, Advanced Telecommunications Research Institute International, 2-2-2 Hikaridai, Seika-cho, Soraku-gun, Kyoto 619-0288, Japan
| | | | | | - Thomas N Sato
- Karydo TherapeutiX, Inc., Kyoto, Japan; ERATO Sato Live Bio-Forecasting Project, Kyoto, Japan; The Thomas N. Sato BioMEC-X Laboratories, Advanced Telecommunications Research Institute International, 2-2-2 Hikaridai, Seika-cho, Soraku-gun, Kyoto 619-0288, Japan; Biomedical Engineering, Cornell University, Ithaca, NY, USA; Centenary Institute, Sydney, Australia; V-iClinix Laboratory, Nara Medical University, Nara, Japan.
| |
Collapse
|
20
|
Ray M, Lakhotia SC. Activated Ras/JNK driven Dilp8 in imaginal discs adversely affects organismal homeostasis during early pupal stage in Drosophila, a new checkpoint for development. Dev Dyn 2019; 248:1211-1231. [PMID: 31415125 DOI: 10.1002/dvdy.102] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 07/23/2019] [Accepted: 08/08/2019] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Dilp8-mediated inhibition of ecdysone synthesis and pupation in holometabolous insects maintains developmental homeostasis through stringent control of timing and strength of molting signals. We examined reasons for normal pupation but early pupal death observed in certain cases. RESULTS Overexpression of activated Ras in developing eye/wing discs inhibited Ptth expression in brain via upregulated JNK signaling mediated Dilp8 secretion from imaginal discs, which inhibited ecdysone synthesis in prothoracic gland after pupariation, leading to death of ~25- to 30-hour-old pupae. Inhibition of elevated Ras signaling completely rescued early pupal death while post-pupation administration of ecdysone to organisms with elevated Ras signaling in eye discs partially rescued their early pupal death. Unlike the earlier known Dilp8 action in delaying pupation, hyperactivated Ras mediated elevation of pJNK signaling in imaginal discs caused Dilp8 secretion after pupariation. Ectopic expression of certain other transgene causing pupal lethality similarly enhanced pJNK and early pupal Dilp8 levels. Suboptimal ecdysone levels after 8 hours of pupation prevented the early pupal metamorphic changes and caused organismal death. CONCLUSIONS Our results reveal early pupal stage as a novel Dilp8 mediated post-pupariation checkpoint and provide further evidence for interorgan signaling during development, wherein a peripheral tissue influences the CNS driven endocrine function.
Collapse
Affiliation(s)
- Mukulika Ray
- Cytogenetics Laboratory, Department of Zoology, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Subhash C Lakhotia
- Cytogenetics Laboratory, Department of Zoology, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| |
Collapse
|
21
|
|
22
|
Imag(in)ing growth and form. Mech Dev 2017; 145:13-21. [DOI: 10.1016/j.mod.2017.03.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 03/21/2017] [Accepted: 03/23/2017] [Indexed: 01/03/2023]
|
23
|
Liu Q, Jin LH. Organ-to-Organ Communication: A Drosophila Gastrointestinal Tract Perspective. Front Cell Dev Biol 2017; 5:29. [PMID: 28421183 PMCID: PMC5376570 DOI: 10.3389/fcell.2017.00029] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 03/15/2017] [Indexed: 01/05/2023] Open
Abstract
The long-term maintenance of an organism's homeostasis and health relies on the accurate regulation of organ-organ communication. Recently, there has been growing interest in using the Drosophila gastrointestinal tract to elucidate the regulatory programs that underlie the complex interactions between organs. Data obtained in this field have dramatically improved our understanding of how organ-organ communication contributes to the regulation of various aspects of the intestine, including its metabolic and physiological status. However, although research uncovering regulatory programs associated with interorgan communication has provided key insights, the underlying mechanisms have not been extensively explored. In this review, we highlight recent findings describing gut-neighbor and neighbor-neighbor communication models in adults and larvae, respectively, with a special focus on how a range of critical strategies concerning continuous interorgan communication and adjustment can be used to manipulate different aspects of biological processes. Given the high degree of similarity between the Drosophila and mammalian intestinal epithelia, it can be anticipated that further analyses of the Drosophila gastrointestinal tract will facilitate the discovery of similar mechanisms underlying organ-organ communication in other mammalian organs, such as the human intestine.
Collapse
Affiliation(s)
- Qiang Liu
- Department of Genetics, College of Life Sciences, Northeast Forestry UniversityHarbin, China
| | - Li Hua Jin
- Department of Genetics, College of Life Sciences, Northeast Forestry UniversityHarbin, China
| |
Collapse
|
24
|
Amourda C, Saunders TE. Gene expression boundary scaling and organ size regulation in the Drosophila embryo. Dev Growth Differ 2017; 59:21-32. [PMID: 28093727 DOI: 10.1111/dgd.12333] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 11/22/2016] [Accepted: 11/23/2016] [Indexed: 12/21/2022]
Abstract
How the shape and size of tissues and organs is regulated during development is a major question in developmental biology. Such regulation relies upon both intrinsic cues (such as signaling networks) and extrinsic inputs (such as from neighboring tissues). Here, we focus on pattern formation and organ development during Drosophila embryogenesis. In particular, we outline the importance of both biochemical and mechanical tissue-tissue interactions in size regulation. We describe how the Drosophila embryo can potentially provide novel insights into how shape and size are regulated during development. We focus on gene expression boundary scaling in the early embryo and how size is regulated in three organs (hindgut, trachea, and ventral nerve cord) later in development, with particular focus on the role of tissue-tissue interactions. Overall, we demonstrate that Drosophila embryogenesis provides a suitable model system for studying spatial and temporal scaling and size control in vivo.
Collapse
Affiliation(s)
- Christopher Amourda
- Mechanobiology Institute, National University of Singapore, T-Lab, #10-01, 5A Engineering Drive 1, 117411, Singapore
| | - Timothy E Saunders
- Mechanobiology Institute, National University of Singapore, T-Lab, #10-01, 5A Engineering Drive 1, 117411, Singapore.,Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, 117543, Singapore.,Institute Of Molecular and Cell Biology, 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| |
Collapse
|
25
|
Abstract
Studies in mammals and Drosophila have demonstrated the existence and significance of secreted factors involved in communication between distal organs. In this review, primarily focusing on Drosophila, we examine the known interorgan communication factors and their functions, physiological inducers, and integration in regulating physiology. Moreover, we describe how organ-sensing screens in Drosophila can systematically identify novel conserved interorgan communication factors. Finally, we discuss how interorgan communication enabled and evolved as a result of specialization of organs. Together, we anticipate that future studies will establish a model for metazoan interorgan communication network (ICN) and how it is deregulated in disease.
Collapse
Affiliation(s)
- Ilia A Droujinine
- Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115; ,
| | - Norbert Perrimon
- Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115; ,
- Howard Hughes Medical Institute, Boston, Massachusetts 02115
| |
Collapse
|
26
|
Roles of the interorgan neuronal network in the development of metabolic syndrome. Diabetol Int 2016; 7:205-211. [PMID: 30603265 DOI: 10.1007/s13340-016-0277-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 06/26/2016] [Indexed: 01/01/2023]
Abstract
Metabolic processes in different tissues and remote organs are under coordinated systemic regulation, allowing adaptation to a variety of external circumstances. Neuronal signals as well as humoral factors, such as nutrients, growth factors, and hormones, have attracted increasing attention for their roles in this interorgan metabolic network, responsible for the maintenance of metabolic homeostasis at the whole-body level. These interorgan communications within an organism are considered to be diverse and, in fact, we identified previously unknown neuronal relay systems originating in the liver which modulate energy, glucose, and lipid metabolism. Furthermore, when nutrient overload is prolonged, these neuronal mechanisms, which function as an endogenous defense system against obesity development, contribute to the pathophysiological states of metabolic syndrome characterized by obesity-associated features. Therefore, these interorgan neuronal systems are considered to be possible molecular targets for treating metabolic syndrome. We herein review the precise mechanisms underlying the functions of the mammalian interorgan neuronal network, especially the pathways from the liver to several other organs, focusing on their significance and roles in the development of metabolic syndrome.
Collapse
|
27
|
Abstract
Several signaling pathways, including the JAK/STAT and Toll pathways, are known to activate blood cells (hemocytes) in Drosophila melanogaster larvae. They are believed to regulate the immune response against infections by parasitoid wasps, such as Leptopilina boulardi, but how these pathways control the hemocytes is not well understood. Here, we discuss the recent discovery that both muscles and fat body take an active part in this response. Parasitoid wasp infection induces Upd2 and Upd3 secretion from hemocytes, leading to JAK/STAT activation mainly in hemocytes and in skeletal muscles. JAK/STAT activation in muscles, but not in hemocytes, is required for an efficient encapsulation of wasp eggs. This suggests that Upd2 and Upd3 are important cytokines, coordinating different tissues for the cellular immune response in Drosophila. In the fat body, Toll signaling initiates a systemic response in which hemocytes are mobilized and activated hemocytes (lamellocytes) are generated. However, the contribution of Toll signaling to the defense against wasps is limited, probably because the wasps inject inhibitors that prevent the activation of the Toll pathway. In conclusion, parasite infection induces a systemic response in Drosophila larvae involving major organ systems and probably the physiology of the entire organism.
Collapse
Affiliation(s)
- Hairu Yang
- a Department of Molecular Biology , Umeå University , Umeå , Sweden
| | - Dan Hultmark
- a Department of Molecular Biology , Umeå University , Umeå , Sweden.,b BioMediTech, University of Tampere , Tampere , Finland
| |
Collapse
|
28
|
Abstract
Many organisms have developed a robust ability to adapt and survive in the face of environmental perturbations that threaten the integrity of their genome, proteome, or metabolome. Studies in multiple model organisms have shown that, in general, when exposed to stress, cells activate a complex prosurvival signaling network that includes immune and DNA damage response genes, chaperones, antioxidant enzymes, structural proteins, metabolic enzymes, and noncoding RNAs. The manner of activation runs the gamut from transcriptional induction of genes to increased stability of transcripts to posttranslational modification of important biosynthetic proteins within the stressed tissue. Superimposed on these largely autonomous effects are nonautonomous responses in which the stressed tissue secretes peptides and other factors that stimulate tissues in different organs to embark on processes that ultimately help the organism as a whole cope with stress. This review focuses on the mechanisms by which tissues in one organ adapt to environmental challenges by regulating stress responses in tissues of different organs.
Collapse
Affiliation(s)
- Edward Owusu-Ansah
- Department of Physiology and Cellular Biophysics, Columbia University Medical Center, New York, NY 10032;
| | | |
Collapse
|
29
|
Goldman AW, Burmeister Y, Cesnulevicius K, Herbert M, Kane M, Lescheid D, McCaffrey T, Schultz M, Seilheimer B, Smit A, St Laurent G, Berman B. Bioregulatory systems medicine: an innovative approach to integrating the science of molecular networks, inflammation, and systems biology with the patient's autoregulatory capacity? Front Physiol 2015; 6:225. [PMID: 26347656 PMCID: PMC4541032 DOI: 10.3389/fphys.2015.00225] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 07/27/2015] [Indexed: 12/25/2022] Open
Abstract
Bioregulatory systems medicine (BrSM) is a paradigm that aims to advance current medical practices. The basic scientific and clinical tenets of this approach embrace an interconnected picture of human health, supported largely by recent advances in systems biology and genomics, and focus on the implications of multi-scale interconnectivity for improving therapeutic approaches to disease. This article introduces the formal incorporation of these scientific and clinical elements into a cohesive theoretical model of the BrSM approach. The authors review this integrated body of knowledge and discuss how the emergent conceptual model offers the medical field a new avenue for extending the armamentarium of current treatment and healthcare, with the ultimate goal of improving population health.
Collapse
Affiliation(s)
- Alyssa W Goldman
- Concept Systems, Inc. Ithaca, NY, USA ; Department of Sociology, Cornell University Ithaca, NY, USA
| | | | | | - Martha Herbert
- Transcend Research Laboratory, Massachusetts General Hospital Boston, MA, USA
| | - Mary Kane
- Concept Systems, Inc. Ithaca, NY, USA
| | - David Lescheid
- International Academy of Bioregulatory Medicine Baden-Baden, Germany
| | - Timothy McCaffrey
- Division of Genomic Medicine, George Washington University Medical Center Washington, DC, USA
| | - Myron Schultz
- Biologische Heilmittel Heel GmbH Baden-Baden, Germany
| | | | - Alta Smit
- Biologische Heilmittel Heel GmbH Baden-Baden, Germany
| | | | - Brian Berman
- Center for Integrative Medicine, University of Maryland School of Medicine Baltimore, MD, USA
| |
Collapse
|
30
|
Schulz AM, Haynes CM. UPR(mt)-mediated cytoprotection and organismal aging. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2015; 1847:1448-56. [PMID: 25857997 DOI: 10.1016/j.bbabio.2015.03.008] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 03/25/2015] [Accepted: 03/28/2015] [Indexed: 12/20/2022]
Abstract
Time- or age-dependent accumulation of mitochondrial damage and dysfunction is strongly associated with aging [1]. Thus, a major biomedical goal is to identify and therapeutically manipulate those inherent programs that protect against mitochondrial dysfunction to promote cell survival and organismal health. The mitochondrial unfolded protein response (UPR(mt)) is such a protective transcriptional response mediated by mitochondrial-to-nuclear signaling that includes mitochondrial proteostasis genes to stabilize mitochondrial function, metabolic adaptations, as well as an innate immunity program. Here, we review the UPR(mt) and its role during a variety of forms of mitochondrial dysfunction including those caused by mutations in respiratory chain genes as well as upon exposure to pathogens that produce mitochondrial toxins. We also review recent data in support of and against the emerging role of the UPR(mt) during aging and longevity. This article is part of a Special Issue entitled: Mitochondrial Dysfunction in Aging.
Collapse
Affiliation(s)
- Anna M Schulz
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA.
| | - Cole M Haynes
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA; BCMB Allied Program, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10065, USA.
| |
Collapse
|
31
|
Apidianakis Y, Ferrandon D. Modeling hologenome imbalances in inflammation and cancer. Front Cell Infect Microbiol 2014; 4:134. [PMID: 25309882 PMCID: PMC4174041 DOI: 10.3389/fcimb.2014.00134] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 09/05/2014] [Indexed: 11/13/2022] Open
Affiliation(s)
| | - Dominique Ferrandon
- UPR9022 CNRS, University of Strasbourg Institute for Advanced Study Strasbourg, France
| |
Collapse
|