1
|
Gergues MM, Lalani LK, Kheirbek MA. Identifying dysfunctional cell types and circuits in animal models for psychiatric disorders with calcium imaging. Neuropsychopharmacology 2024; 50:274-284. [PMID: 39122815 PMCID: PMC11525937 DOI: 10.1038/s41386-024-01942-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 05/30/2024] [Accepted: 07/09/2024] [Indexed: 08/12/2024]
Abstract
A central goal of neuroscience is to understand how the brain transforms external stimuli and internal bodily signals into patterns of activity that underlie cognition, emotional states, and behavior. Understanding how these patterns of activity may be disrupted in mental illness is crucial for developing novel therapeutics. It is well appreciated that psychiatric disorders are complex, circuit-based disorders that arise from dysfunctional activity patterns generated in discrete cell types and their connections. Recent advances in large-scale, cell-type specific calcium imaging approaches have shed new light on the cellular, circuit, and network-level dysfunction in animal models for psychiatric disorders. Here, we highlight a series of recent findings over the last ~10 years from in vivo calcium imaging studies that show how aberrant patterns of activity in discrete cell types and circuits may underlie behavioral deficits in animal models for several psychiatric disorders, including depression, anxiety, autism spectrum disorders, and schizophrenia. These advances in calcium imaging in pre-clinical models demonstrate the power of cell-type-specific imaging tools in understanding the underlying dysfunction in cell types, activity patterns, and neural circuits that may contribute to disease and provide new blueprints for developing more targeted therapeutics and treatment strategies.
Collapse
Affiliation(s)
- Mark M Gergues
- Neuroscience Graduate Program, University of California San Francisco, San Francisco, CA, USA
- Department of Psychiatry and Behavioral Sciences, University of California San Francisco, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Lahin K Lalani
- Neuroscience Graduate Program, University of California San Francisco, San Francisco, CA, USA
- Department of Psychiatry and Behavioral Sciences, University of California San Francisco, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Mazen A Kheirbek
- Neuroscience Graduate Program, University of California San Francisco, San Francisco, CA, USA.
- Department of Psychiatry and Behavioral Sciences, University of California San Francisco, San Francisco, CA, USA.
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA.
- Kavli Institute for Fundamental Neuroscience, University of California San Francisco, San Francisco, CA, USA.
- Center for Integrative Neuroscience, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
2
|
Kumar S, Bhushan B, Kumar A, Panigrahi M, Bharati J, Kumari S, Kaiho K, Banik S, Karthikeyan A, Chaudhary R, Gaur GK, Dutt T. Elucidation of novel SNPs affecting immune response to classical swine fever vaccination in pigs using immunogenomics approach. Vet Res Commun 2024; 48:941-953. [PMID: 38017322 DOI: 10.1007/s11259-023-10262-3] [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: 09/11/2023] [Accepted: 11/19/2023] [Indexed: 11/30/2023]
Abstract
The host genetic makeup plays a significant role in causing the within-breed variation among individuals after vaccination. The present study was undertaken to elucidate the genetic basis of differential immune response between high and low responder Landlly (Landrace X Ghurrah) piglets vis-à-vis CSF vaccination. For the purpose, E2 antibody response against CSF vaccination was estimated in sampled animals on the day of vaccination and 21-day post-vaccination as a measure of humoral immune response. Double-digestion restriction associated DNA (ddRAD) sequencing was undertaken on 96 randomly chosen Landlly piglets using Illumina HiSeq platform. SNP markers were called using standard methodology. Genome-wide association study (GWAS) was undertaken in PLINK program to identify the informative SNP markers significantly associated with differential immune response. The results revealed significant SNPs associated with E2 antibody response against CSF vaccination. The genome-wide informative SNPs for the humoral immune response against CSF vaccination were located on SSC10, SSC17, SSC9, SSC2, SSC3 and SSC6. The overlapping and flanking genes (500Kb upstream and downstream) of significant SNPs were CYB5R1, PCMTD2, WT1, IL9R, CD101, TMEM64, TLR6, PIGG, ADIPOR1, PRSS37, EIF3M, and DNAJC24. Functional enrichment and annotation analysis were undertaken for these genes in order to gain maximum insights into the association of these genes with immune system functionality in pigs. The genetic makeup was associated with differential immune response against CSF vaccination in Landlly piglets while the identified informative SNPs may be used as suitable markers for determining variation in host immune response against CSF vaccination in pigs.
Collapse
Affiliation(s)
- Satish Kumar
- Division of Animal Genetics, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, U.P, 243122, India.
- ICAR-National Research Centre on Pig, Rani, Guwahati, Assam, 781131, India.
| | - Bharat Bhushan
- Division of Animal Genetics, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, U.P, 243122, India.
| | - Amit Kumar
- Division of Animal Genetics, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, U.P, 243122, India.
| | - Manjit Panigrahi
- Division of Animal Genetics, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, U.P, 243122, India
| | - Jaya Bharati
- ICAR-National Research Centre on Pig, Rani, Guwahati, Assam, 781131, India
| | - Soni Kumari
- Division of Animal Genetics, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, U.P, 243122, India
| | - Kaisa Kaiho
- Division of Animal Genetics, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, U.P, 243122, India
| | - Santanu Banik
- ICAR-National Research Centre on Pig, Rani, Guwahati, Assam, 781131, India
| | - A Karthikeyan
- Division of Animal Genetics, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, U.P, 243122, India
| | - Rajni Chaudhary
- Division of Animal Genetics, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, U.P, 243122, India
| | - G K Gaur
- Livestock Production and Management Section, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, U.P, 243122, India
| | - Triveni Dutt
- Livestock Production and Management Section, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, U.P, 243122, India
| |
Collapse
|
3
|
Bohlson SS, Tenner AJ. Complement in the Brain: Contributions to Neuroprotection, Neuronal Plasticity, and Neuroinflammation. Annu Rev Immunol 2023; 41:431-452. [PMID: 36750318 DOI: 10.1146/annurev-immunol-101921-035639] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
The complement system is an ancient collection of proteolytic cascades with well-described roles in regulation of innate and adaptive immunity. With the convergence of a revolution in complement-directed clinical therapeutics, the discovery of specific complement-associated targetable pathways in the central nervous system, and the development of integrated multi-omic technologies that have all emerged over the last 15 years, precision therapeutic targeting in Alzheimer disease and other neurodegenerative diseases and processes appears to be within reach. As a sensor of tissue distress, the complement system protects the brain from microbial challenge as well as the accumulation of dead and/or damaged molecules and cells. Additional more recently discovered diverse functions of complement make it of paramount importance to design complement-directed neurotherapeutics such that the beneficial roles in neurodevelopment, adult neural plasticity, and neuroprotective functions of the complement system are retained.
Collapse
Affiliation(s)
- Suzanne S Bohlson
- Department of Molecular Biology and Biochemistry, University of California, Irvine, California, USA; ,
| | - Andrea J Tenner
- Department of Molecular Biology and Biochemistry, University of California, Irvine, California, USA; ,
- Department of Neurobiology and Behavior, University of California, Irvine, California, USA
- Department of Pathology and Laboratory Medicine, School of Medicine, University of California, Irvine, California, USA
| |
Collapse
|
4
|
Papadakos KS, Ekström A, Slipek P, Skourti E, Reid S, Pietras K, Blom AM. Sushi domain-containing protein 4 binds to epithelial growth factor receptor and initiates autophagy in an EGFR phosphorylation independent manner. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2022; 41:363. [PMID: 36578014 PMCID: PMC9798675 DOI: 10.1186/s13046-022-02565-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 12/07/2022] [Indexed: 12/29/2022]
Abstract
BACKGROUND Sushi domain-containing protein 4 (SUSD4) is a recently discovered protein with unknown cellular functions. We previously revealed that SUSD4 can act as complement inhibitor and as a potential tumor suppressor. METHODS In a syngeneic mouse model of breast cancer, tumors expressing SUSD4 had a smaller volume compared with the corresponding mock control tumors. Additionally, data from three different expression databases and online analysis tools confirm that for breast cancer patients, high mRNA expression of SUSD4 in the tumor tissue correlates with a better prognosis. In vitro experiments utilized triple-negative breast cancer cell lines (BT-20 and MDA-MB-468) stably expressing SUSD4. Moreover, we established a cell line based on BT-20 in which the gene for EGFR was knocked out with the CRISPR-Cas9 method. RESULTS We discovered that the Epithelial Growth Factor Receptor (EGFR) interacts with SUSD4. Furthermore, triple-negative breast cancer cell lines stably expressing SUSD4 had higher autophagic flux. The initiation of autophagy required the expression of EGFR but not phosphorylation of the receptor. Expression of SUSD4 in the breast cancer cells led to activation of the tumor suppressor LKB1 and consequently to the activation of AMPKα1. Finally, autophagy was initiated after stimulation of the ULK1, Atg14 and Beclin-1 axis in SUSD4 expressing cells. CONCLUSIONS In this study we provide novel insight into the molecular mechanism of action whereby SUSD4 acts as an EGFR inhibitor without affecting the phosphorylation of the receptor and may potentially influence the recycling of EGFR to the plasma membrane.
Collapse
Affiliation(s)
- Konstantinos S. Papadakos
- grid.4514.40000 0001 0930 2361Division of Medical Protein Chemistry, Department of Translational Medicine, Lund University, Inga Maria Nilsson’s street 53, 214 28 Malmö, Sweden
| | - Alexander Ekström
- grid.4514.40000 0001 0930 2361Division of Medical Protein Chemistry, Department of Translational Medicine, Lund University, Inga Maria Nilsson’s street 53, 214 28 Malmö, Sweden
| | - Piotr Slipek
- grid.4514.40000 0001 0930 2361Division of Medical Protein Chemistry, Department of Translational Medicine, Lund University, Inga Maria Nilsson’s street 53, 214 28 Malmö, Sweden
| | - Eleni Skourti
- grid.4514.40000 0001 0930 2361Division of Medical Protein Chemistry, Department of Translational Medicine, Lund University, Inga Maria Nilsson’s street 53, 214 28 Malmö, Sweden
| | - Steven Reid
- grid.4514.40000 0001 0930 2361Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Kristian Pietras
- grid.4514.40000 0001 0930 2361Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Anna M. Blom
- grid.4514.40000 0001 0930 2361Division of Medical Protein Chemistry, Department of Translational Medicine, Lund University, Inga Maria Nilsson’s street 53, 214 28 Malmö, Sweden
| |
Collapse
|
5
|
Pryzdial ELG, Leatherdale A, Conway EM. Coagulation and complement: Key innate defense participants in a seamless web. Front Immunol 2022; 13:918775. [PMID: 36016942 PMCID: PMC9398469 DOI: 10.3389/fimmu.2022.918775] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 07/06/2022] [Indexed: 12/30/2022] Open
Abstract
In 1969, Dr. Oscar Ratnoff, a pioneer in delineating the mechanisms by which coagulation is activated and complement is regulated, wrote, “In the study of biological processes, the accumulation of information is often accelerated by a narrow point of view. The fastest way to investigate the body’s defenses against injury is to look individually at such isolated questions as how the blood clots or how complement works. We must constantly remind ourselves that such distinctions are man-made. In life, as in the legal cliché, the devices through which the body protects itself form a seamless web, unwrinkled by our artificialities.” Our aim in this review, is to highlight the critical molecular and cellular interactions between coagulation and complement, and how these two major component proteolytic pathways contribute to the seamless web of innate mechanisms that the body uses to protect itself from injury, invading pathogens and foreign surfaces.
Collapse
Affiliation(s)
- Edward L. G. Pryzdial
- Centre for Blood Research, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
- Canadian Blood Services, Medical Affairs and Innovation, Vancouver, BC, Canada
- *Correspondence: Edward L. G. Pryzdial, ; Edward M. Conway,
| | - Alexander Leatherdale
- Centre for Blood Research, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
- Division of Hematology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Edward M. Conway
- Centre for Blood Research, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
- Canadian Blood Services, Medical Affairs and Innovation, Vancouver, BC, Canada
- Division of Hematology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
- *Correspondence: Edward L. G. Pryzdial, ; Edward M. Conway,
| |
Collapse
|
6
|
Rios-Barros LV, Silva-Moreira AL, Horta MF, Gontijo NF, Castro-Gomes T. How to get away with murder: The multiple strategies employed by pathogenic protozoa to avoid complement killing. Mol Immunol 2022; 149:27-38. [PMID: 35709630 DOI: 10.1016/j.molimm.2022.05.118] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 05/16/2022] [Accepted: 05/24/2022] [Indexed: 01/15/2023]
Abstract
Parasitic protozoa are eukaryotic unicellular organisms that depend on a variety of living organisms and can develop intra- and extracellularly inside their hosts. In humans, these parasites cause diseases with a significant impact on public health, such as malaria, toxoplasmosis, Chagas disease, leishmaniasis and amebiasis. The ability of a parasite in establishing a successful infection depends on a series of intricate evolutionarily selected adaptations, which include the development of molecular and cellular strategies to evade the host immune system effector mechanisms. The complement system is one of the main effector mechanisms and the first humoral shield of hosts innate immunity against pathogens. For unicellular pathogens, such as protozoa, bacteria and fungi, the activation of the complement system may culminate in the elimination of the invader mainly via 1- the formation of a pore that depolarizes the plasma membrane of the parasite, causing cell lysis; 2- opsonization and killing by phagocytes; 3- increasing vascular permeability while also recruiting neutrophils to the site of activation. Numerous strategies to avoid complement activation have been reported for parasitic protozoa, such as 1- sequestration of complement system regulatory proteins produced by the host, 2- expression of complement system regulatory proteins, 3- proteolytic cleavage of different complement effector molecules, 4- formation of a physical glycolipid barrier that prevents deposition of complement molecules on the plasma membrane, and 5- removal, by endocytosis, of complement molecules bound to plasma membrane. In this review, we revisit the different strategies of blocking various stages of complement activation described for the main species of parasitic protozoa, present the most recent discoveries in the field and discuss new perspectives on yet neglected strategies and possible new evasion mechanisms.
Collapse
Affiliation(s)
- Laura Valeria Rios-Barros
- Departamento de Parasitologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil.
| | - Anna Luiza Silva-Moreira
- Departamento de Parasitologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil.
| | - Maria Fatima Horta
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil.
| | - Nelder Figueiredo Gontijo
- Departamento de Parasitologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil.
| | - Thiago Castro-Gomes
- Departamento de Parasitologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
7
|
Parker SE, Bellingham MC, Woodruff TM. Complement drives circuit modulation in the adult brain. Prog Neurobiol 2022; 214:102282. [DOI: 10.1016/j.pneurobio.2022.102282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 03/24/2022] [Accepted: 05/02/2022] [Indexed: 11/16/2022]
|
8
|
Song W, Li Q, Wang T, Li Y, Fan T, Zhang J, Wang Q, Pan J, Dong Q, Sun ZS, Wang Y. Putative complement control protein CSMD3 dysfunction impairs synaptogenesis and induces neurodevelopmental disorders. Brain Behav Immun 2022; 102:237-250. [PMID: 35245678 DOI: 10.1016/j.bbi.2022.02.027] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 02/10/2022] [Accepted: 02/26/2022] [Indexed: 12/23/2022] Open
Abstract
Recent studies have reported that complement-related proteins modulate brain development through regulating synapse processes in the cortex. CSMD3 belongs to a group of putative complement control proteins. However, its role in the central nervous system and synaptogenesis remains largely unknown. Here we report that CSMD3 deleterious mutations occur frequently in patients with neurodevelopmental disorders (NDDs). Csmd3 is predominantly expressed in cortical neurons of the developing cortex. In mice, Csmd3 disruption induced retarded development and NDD-related behaviors. Csmd3 deficiency impaired synaptogenesis and neurogenesis, allowing fewer neurons reaching the cortical plate. Csmd3 deficiency also induced perturbed functional networks in the developing cortex, involving a number of downregulated synapse-associated genes that influence early synaptic organization and upregulated genes related to immune activity. Our study provides mechanistic insights into the endogenous regulation of complement-related proteins in synaptic development and supports the pathological role of CSMD3 in NDDs.
Collapse
Affiliation(s)
- Wei Song
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, China; CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Quan Li
- School of Life Sciences, Hebei University, Baoding 071002, China
| | - Tao Wang
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, China
| | - Yuanyuan Li
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, China; CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Tianda Fan
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou 325000, China
| | - Jianghong Zhang
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, China; CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qingqing Wang
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jinrong Pan
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, China; CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qiwen Dong
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhong Sheng Sun
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, China; CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing 100049, China; School of Life Sciences, Hebei University, Baoding 071002, China; Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou 325000, China; State Key Laboratory of Integrated Management of Pest Insects and Rodents, Chinese Academy of Sciences, Beijing 100101, China.
| | - Yan Wang
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, China; CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
9
|
Qiu Y, Shen S, Yang Y, Wang W. An Excretory Protein of Echinococcus multilocularis Inhibits Complement Classical Pathway Activation. Infect Drug Resist 2022; 15:555-568. [PMID: 35228806 PMCID: PMC8881923 DOI: 10.2147/idr.s344075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 02/07/2022] [Indexed: 11/23/2022] Open
Abstract
Introduction Alveolar echinococcosis is a lethal zoonosis caused by Echinococcus multilocularis (E.m) larvae. The mechanism by which E.m evades host immune attacks and ensures long-term survival remains unexplained. The complement system is a cascade of sequentially activated complement proteins that results in opsonization-related phagocytosis or membrane lysis of invading organisms. Excretory/secretory proteins (ESPs) of parasites are the main antigens that induce the immune response and play important roles in the long-term survival. Methods We investigated the possibility that E.m inhibits complement activation through ESPs and examined the potential related mechanism. A haemolysis assay was used to determine if and how in vitro culture medium of E.m containing ESPs can inhibit complement activation. Potential ESPs were annotated using bioinformatics methods, and one ESP was subsequently expressed as a recombinant protein with a eukaryotic expression system. The ability of this protein to inhibit complement activation was also tested by haemolysis assay. Results These assays showed that in vitro culture medium of E.m inhibited activation of the complement classical pathway. EmuJ_000439500 encodes a protein containing seven Sushi domains, which was the only potential E.m-derived complement inhibitor (Em-CI, UniProt: A0A068Y4F2) annotated among the 653 ESPs. Recombinant Em-CI also displayed the ability to inhibit activation of the complement classical pathway. Discussion The discovery of Em-CI sheds light on the mechanism by which E.m escapes killing by the complement system and provides potential targets for immunotherapy for parasitic diseases.
Collapse
Affiliation(s)
- Yiwen Qiu
- Department of Liver Surgery and Liver Transplantation Center, West China Hospital of Sichuan University, Chengdu, People’s Republic of China
| | - Shu Shen
- Department of Liver Surgery and Liver Transplantation Center, West China Hospital of Sichuan University, Chengdu, People’s Republic of China
| | - Yi Yang
- Department of Liver Surgery and Liver Transplantation Center, West China Hospital of Sichuan University, Chengdu, People’s Republic of China
| | - Wentao Wang
- Department of Liver Surgery and Liver Transplantation Center, West China Hospital of Sichuan University, Chengdu, People’s Republic of China
- Correspondence: Wentao Wang, Department of Liver Surgery and Liver Transplantation Center, West China Hospital of Sichuan University, 37 Guoxue Road, Chengdu, 610041, People’s Republic of China, Tel +86 18980601895, Fax +86-028-85422871, Email
| |
Collapse
|
10
|
C-type lectin binds envelope protein of white spot syndrome virus and induces antiviral peptides in red swamp crayfish. FISH AND SHELLFISH IMMUNOLOGY REPORTS 2021; 2:100027. [DOI: 10.1016/j.fsirep.2021.100027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 09/04/2021] [Accepted: 09/08/2021] [Indexed: 10/20/2022] Open
|
11
|
Complement as a powerful "influencer" in the brain during development, adulthood and neurological disorders. Adv Immunol 2021; 152:157-222. [PMID: 34844709 DOI: 10.1016/bs.ai.2021.09.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The complement system was long considered as only a powerful effector arm of the immune system that, while critically protective, could lead to inflammation and cell death if overactivated, even in the central nervous system (CNS). However, in the past decade it has been recognized as playing critical roles in key physiological processes in the CNS, including neurogenesis and synaptic remodeling in the developing and adult brain. Inherent in these processes are the interactions with cells in the brain, and the cascade of interactions and functional consequences that ensue. As a result, investigations of therapeutic approaches for both suppressing excessive complement driven neurotoxicity and aberrant sculpting of neuronal circuits, require broad (and deep) knowledge of the functional activities of multiple components of this highly evolved and regulated system to avoid unintended negative consequences in the clinic. Advances in basic science are beginning to provide a roadmap for translation to therapeutics, with both small molecule and biologics. Here, we present examples of the critical roles of proper complement function in the development and sculpting of the nervous system, and in enabling rapid protection from infection and clearance of dying cells. Microglia are highlighted as important command centers that integrate signals from the complement system and other innate sensors that are programed to provide support and protection, but that direct detrimental responses to aberrant activation and/or regulation of the system. Finally, we present promising research areas that may lead to effective and precision strategies for complement targeted interventions to promote neurological health.
Collapse
|
12
|
Qiu WQ, Luo S, Ma SA, Saminathan P, Li H, Gunnersen JM, Gelbard HA, Hammond JW. The Sez6 Family Inhibits Complement by Facilitating Factor I Cleavage of C3b and Accelerating the Decay of C3 Convertases. Front Immunol 2021; 12:607641. [PMID: 33936031 PMCID: PMC8081827 DOI: 10.3389/fimmu.2021.607641] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 03/23/2021] [Indexed: 12/31/2022] Open
Abstract
The Sez6 family consists of Sez6, Sez6L, and Sez6L2. Its members are expressed throughout the brain and have been shown to influence synapse numbers and dendritic morphology. They are also linked to various neurological and psychiatric disorders. All Sez6 family members contain 2-3 CUB domains and 5 complement control protein (CCP) domains, suggesting that they may be involved in complement regulation. We show that Sez6 family members inhibit C3b/iC3b opsonization by the classical and alternative pathways with varying degrees of efficacy. For the classical pathway, Sez6 is a strong inhibitor, Sez6L2 is a moderate inhibitor, and Sez6L is a weak inhibitor. For the alternative pathway, the complement inhibitory activity of Sez6, Sez6L, and Sez6L2 all equaled or exceeded the activity of the known complement regulator MCP. Using Sez6L2 as the representative family member, we show that it specifically accelerates the dissociation of C3 convertases. Sez6L2 also functions as a cofactor for Factor I to facilitate the cleavage of C3b; however, Sez6L2 has no cofactor activity toward C4b. In summary, the Sez6 family are novel complement regulators that inhibit C3 convertases and promote C3b degradation.
Collapse
Affiliation(s)
- Wen Q Qiu
- Center for Neurotherapeutics Discovery, Department of Neurology, University of Rochester Medical Center, Rochester, NY, United States
| | - Shaopeiwen Luo
- Center for Neurotherapeutics Discovery, Department of Neurology, University of Rochester Medical Center, Rochester, NY, United States
| | - Stefanie A Ma
- Center for Neurotherapeutics Discovery, Department of Neurology, University of Rochester Medical Center, Rochester, NY, United States
| | - Priyanka Saminathan
- Center for Neurotherapeutics Discovery, Department of Neurology, University of Rochester Medical Center, Rochester, NY, United States
| | - Herman Li
- Center for Neurotherapeutics Discovery, Department of Neurology, University of Rochester Medical Center, Rochester, NY, United States
| | - Jenny M Gunnersen
- Department of Anatomy and Neuroscience and The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, Australia
| | - Harris A Gelbard
- Center for Neurotherapeutics Discovery, Department of Neurology, University of Rochester Medical Center, Rochester, NY, United States
| | - Jennetta W Hammond
- Center for Neurotherapeutics Discovery, Department of Neurology, University of Rochester Medical Center, Rochester, NY, United States
| |
Collapse
|
13
|
González-Calvo I, Iyer K, Carquin M, Khayachi A, Giuliani FA, Sigoillot SM, Vincent J, Séveno M, Veleanu M, Tahraoui S, Albert M, Vigy O, Bosso-Lefèvre C, Nadjar Y, Dumoulin A, Triller A, Bessereau JL, Rondi-Reig L, Isope P, Selimi F. Sushi domain-containing protein 4 controls synaptic plasticity and motor learning. eLife 2021; 10:65712. [PMID: 33661101 PMCID: PMC7972451 DOI: 10.7554/elife.65712] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 03/03/2021] [Indexed: 01/28/2023] Open
Abstract
Fine control of protein stoichiometry at synapses underlies brain function and plasticity. How proteostasis is controlled independently for each type of synaptic protein in a synapse-specific and activity-dependent manner remains unclear. Here, we show that Susd4, a gene coding for a complement-related transmembrane protein, is expressed by many neuronal populations starting at the time of synapse formation. Constitutive loss-of-function of Susd4 in the mouse impairs motor coordination adaptation and learning, prevents long-term depression at cerebellar synapses, and leads to misregulation of activity-dependent AMPA receptor subunit GluA2 degradation. We identified several proteins with known roles in the regulation of AMPA receptor turnover, in particular ubiquitin ligases of the NEDD4 subfamily, as SUSD4 binding partners. Our findings shed light on the potential role of SUSD4 mutations in neurodevelopmental diseases.
Collapse
Affiliation(s)
- Inés González-Calvo
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS, INSERM, PSL Research University, Paris, France.,Institut des Neurosciences Cellulaires et Intégratives (INCI), CNRS, Université de Strasbourg, Strasbourg, France
| | - Keerthana Iyer
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS, INSERM, PSL Research University, Paris, France
| | - Mélanie Carquin
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS, INSERM, PSL Research University, Paris, France
| | - Anouar Khayachi
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS, INSERM, PSL Research University, Paris, France
| | - Fernando A Giuliani
- Institut des Neurosciences Cellulaires et Intégratives (INCI), CNRS, Université de Strasbourg, Strasbourg, France
| | - Séverine M Sigoillot
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS, INSERM, PSL Research University, Paris, France
| | - Jean Vincent
- Institut Biology Paris Seine (IBPS), Neuroscience Paris Seine (NPS), CeZaMe, CNRS, Sorbonne University, INSERM, Paris, France
| | - Martial Séveno
- BioCampus Montpellier, CNRS, INSERM, Université de Montpellier, Montpellier, France
| | - Maxime Veleanu
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS, INSERM, PSL Research University, Paris, France
| | - Sylvana Tahraoui
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS, INSERM, PSL Research University, Paris, France
| | - Mélanie Albert
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS, INSERM, PSL Research University, Paris, France
| | - Oana Vigy
- Institut de Génomique Fonctionnelle, CNRS, INSERM, Université de Montpellier, Montpellier, France
| | - Célia Bosso-Lefèvre
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS, INSERM, PSL Research University, Paris, France
| | - Yann Nadjar
- École Normale Supérieure, Institut de Biologie de l'ENS, INSERM, CNRS, PSL Research University, Paris, France
| | - Andréa Dumoulin
- École Normale Supérieure, Institut de Biologie de l'ENS, INSERM, CNRS, PSL Research University, Paris, France
| | - Antoine Triller
- École Normale Supérieure, Institut de Biologie de l'ENS, INSERM, CNRS, PSL Research University, Paris, France
| | - Jean-Louis Bessereau
- Université de Lyon, Université Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U 1217, Institut Neuromyogène, Lyon, France
| | - Laure Rondi-Reig
- Institut Biology Paris Seine (IBPS), Neuroscience Paris Seine (NPS), CeZaMe, CNRS, Sorbonne University, INSERM, Paris, France
| | - Philippe Isope
- Institut des Neurosciences Cellulaires et Intégratives (INCI), CNRS, Université de Strasbourg, Strasbourg, France
| | - Fekrije Selimi
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS, INSERM, PSL Research University, Paris, France
| |
Collapse
|
14
|
More than a Pore: Nonlytic Antimicrobial Functions of Complement and Bacterial Strategies for Evasion. Microbiol Mol Biol Rev 2021; 85:85/1/e00177-20. [PMID: 33504655 DOI: 10.1128/mmbr.00177-20] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The complement system is an evolutionarily ancient defense mechanism against foreign substances. Consisting of three proteolytic activation pathways, complement converges on a common effector cascade terminating in the formation of a lytic pore on the target surface. The classical and lectin pathways are initiated by pattern recognition molecules binding to specific ligands, while the alternative pathway is constitutively active at low levels in circulation. Complement-mediated killing is essential for defense against many Gram-negative bacterial pathogens, and genetic deficiencies in complement can render individuals highly susceptible to infection, for example, invasive meningococcal disease. In contrast, Gram-positive bacteria are inherently resistant to the direct bactericidal activity of complement due to their thick layer of cell wall peptidoglycan. However, complement also serves diverse roles in immune defense against all bacteria by flagging them for opsonization and killing by professional phagocytes, synergizing with neutrophils, modulating inflammatory responses, regulating T cell development, and cross talk with coagulation cascades. In this review, we discuss newly appreciated roles for complement beyond direct membrane lysis, incorporate nonlytic roles of complement into immunological paradigms of host-pathogen interactions, and identify bacterial strategies for complement evasion.
Collapse
|
15
|
Cong Q, Soteros BM, Wollet M, Kim JH, Sia GM. The endogenous neuronal complement inhibitor SRPX2 protects against complement-mediated synapse elimination during development. Nat Neurosci 2020; 23:1067-1078. [PMID: 32661396 PMCID: PMC7483802 DOI: 10.1038/s41593-020-0672-0] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 06/16/2020] [Indexed: 02/06/2023]
Abstract
Complement-mediated synapse elimination has emerged as an important process in both brain development and neurological diseases, but whether neurons express complement inhibitors that protect synapses against complement-mediated synapse elimination remains unknown. Here, we show that the sushi domain protein SRPX2 is a neuronally expressed complement inhibitor that regulates complement-dependent synapse elimination. SRPX2 directly binds to C1q and blocks its activity, and SRPX2-/Y mice show increased C3 deposition and microglial synapse engulfment. They also show a transient decrease in synapse numbers and increase in retinogeniculate axon segregation in the lateral geniculate nucleus. In the somatosensory cortex, SRPX2-/Y mice show decreased thalamocortical synapse numbers and increased spine pruning. C3-/-;SRPX2-/Y double-knockout mice exhibit phenotypes associated with C3-/- mice rather than SRPX2-/Y mice, which indicates that C3 is necessary for the effect of SRPX2 on synapse elimination. Together, these results show that SRPX2 protects synapses against complement-mediated elimination in both the thalamus and the cortex.
Collapse
Affiliation(s)
- Qifei Cong
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Breeanne M Soteros
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Mackenna Wollet
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Jun Hee Kim
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Gek-Ming Sia
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
| |
Collapse
|
16
|
Zhou RR, Wang L, Zhao SD. Estimation and inference for the indirect effect in high-dimensional linear mediation models. Biometrika 2020; 107:573-589. [PMID: 32831353 DOI: 10.1093/biomet/asaa016] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Indexed: 12/19/2022] Open
Abstract
Mediation analysis is difficult when the number of potential mediators is larger than the sample size. In this paper we propose new inference procedures for the indirect effect in the presence of high-dimensional mediators for linear mediation models. We develop methods for both incomplete mediation, where a direct effect may exist, and complete mediation, where the direct effect is known to be absent. We prove consistency and asymptotic normality of our indirect effect estimators. Under complete mediation, where the indirect effect is equivalent to the total effect, we further prove that our approach gives a more powerful test compared to directly testing for the total effect. We confirm our theoretical results in simulations, as well as in an integrative analysis of gene expression and genotype data from a pharmacogenomic study of drug response. We present a novel analysis of gene sets to understand the molecular mechanisms of drug response, and also identify a genome-wide significant noncoding genetic variant that cannot be detected using standard analysis methods.
Collapse
Affiliation(s)
- Ruixuan Rachel Zhou
- Department of Statistics, University of Illinois at Urbana-Champaign, 725 S. Wright Street, Champaign, Illinois 61820, U.S.A
| | - Liewei Wang
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First St. SW, Rochester, Minnesota 55905, U.S.A
| | - Sihai Dave Zhao
- Department of Statistics, University of Illinois at Urbana-Champaign, 725 S. Wright Street, Champaign, Illinois 61820, U.S.A
| |
Collapse
|
17
|
Zhu H, Meissner LE, Byrnes C, Tuymetova G, Tifft CJ, Proia RL. The Complement Regulator Susd4 Influences Nervous-System Function and Neuronal Morphology in Mice. iScience 2020; 23:100957. [PMID: 32179479 PMCID: PMC7075988 DOI: 10.1016/j.isci.2020.100957] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 10/31/2019] [Accepted: 02/25/2020] [Indexed: 01/02/2023] Open
Abstract
The SUSD4 (Sushi domain-containing protein 4) gene encodes a complement inhibitor that is frequently deleted in 1q41q42 microdeletion syndrome, a multisystem congenital disorder that includes neurodevelopmental abnormalities. To understand SUSD4's role in the mammalian nervous system, we analyzed Susd4 knockout (KO) mice. Susd4 KO mice exhibited significant defects in motor performance and significantly higher levels of anxiety-like behaviors. Susd4 KO brain had abnormal “hairy” basket cells surrounding Purkinje neurons within the cerebellum and significantly reduced dendritic spine density in hippocampal pyramidal neurons. Neurons and oligodendrocyte lineage cells of wild-type mice were found to express Susd4 mRNA. Protein expression of the complement component C1q was increased in the brains of Susd4 KO mice. Our data indicate that SUSD4 plays an important role in neuronal functions, possibly via the complement pathway, and that SUSD4 deletion may contribute to the nervous system abnormalities in patients with 1q41q42 deletions. Susd4 is expressed in neurons and oligodendrocyte lineage cells Susd4 knockout mice have abnormal hippocampal and cerebellar neuronal morphologies Susd4 knockout mice exhibit anxiety-like behaviors and impaired motor function Susd4 knockout mice have elevated brain levels of the complement component C1q
Collapse
Affiliation(s)
- Hongling Zhu
- Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD 20892, USA
| | - Laura E Meissner
- Office of the Clinical Director and Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Colleen Byrnes
- Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD 20892, USA
| | - Galina Tuymetova
- Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD 20892, USA
| | - Cynthia J Tifft
- Office of the Clinical Director and Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Richard L Proia
- Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD 20892, USA.
| |
Collapse
|
18
|
Ghallab A, Myllys M, H. Holland C, Zaza A, Murad W, Hassan R, A. Ahmed Y, Abbas T, A. Abdelrahim E, Schneider KM, Matz-Soja M, Reinders J, Gebhardt R, Berres ML, Hatting M, Drasdo D, Saez-Rodriguez J, Trautwein C, G. Hengstler J. Influence of Liver Fibrosis on Lobular Zonation. Cells 2019; 8:E1556. [PMID: 31810365 PMCID: PMC6953125 DOI: 10.3390/cells8121556] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 11/25/2019] [Accepted: 11/28/2019] [Indexed: 12/18/2022] Open
Abstract
Little is known about how liver fibrosis influences lobular zonation. To address this question, we used three mouse models of liver fibrosis, repeated CCl4 administration for 2, 6 and 12 months to induce pericentral damage, as well as bile duct ligation (21 days) and mdr2-/- mice to study periportal fibrosis. Analyses were performed by RNA-sequencing, immunostaining of zonated proteins and image analysis. RNA-sequencing demonstrated a significant enrichment of pericentral genes among genes downregulated by CCl4; vice versa, periportal genes were enriched among the upregulated genes. Immunostaining showed an almost complete loss of pericentral proteins, such as cytochrome P450 enzymes and glutamine synthetase, while periportal proteins, such as arginase 1 and CPS1 became expressed also in pericentral hepatocytes. This pattern of fibrosis-associated 'periportalization' was consistently observed in all three mouse models and led to complete resistance to hepatotoxic doses of acetaminophen (200 mg/kg). Characterization of the expression response identified the inflammatory pathways TGFβ, NFκB, TNFα, and transcription factors NFKb1, Stat1, Hif1a, Trp53, and Atf1 among those activated, while estrogen-associated pathways, Hnf4a and Hnf1a, were decreased. In conclusion, liver fibrosis leads to strong alterations of lobular zonation, where the pericentral region adopts periportal features. Beside adverse consequences, periportalization supports adaptation to repeated doses of hepatotoxic compounds.
Collapse
Affiliation(s)
- Ahmed Ghallab
- Leibniz Research Centre for Working Environment and Human Factors at the Technical University Dortmund, 44139 Dortmund, Germany, (A.Z.); , (J.R.); (D.D.)
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, South Valley University, Qena 83523, Egypt
| | - Maiju Myllys
- Leibniz Research Centre for Working Environment and Human Factors at the Technical University Dortmund, 44139 Dortmund, Germany, (A.Z.); , (J.R.); (D.D.)
| | - Christian H. Holland
- Faculty of Medicine, Institute of Computational Biomedicine, Heidelberg University, Bioquant—Im Neuenheimer Feld 267, 69120 Heidelberg, Germany; (C.H.H.); (J.S.-R.)
- Faculty of Medicine, Joint Research Centre for Computational Biomedicine (JRC-COMBINE), RWTH Aachen University, Pauwelsstrasse 19, 52074 Aachen, Germany
| | - Ayham Zaza
- Leibniz Research Centre for Working Environment and Human Factors at the Technical University Dortmund, 44139 Dortmund, Germany, (A.Z.); , (J.R.); (D.D.)
| | - Walaa Murad
- Histology Department, Faculty of Medicine, South Valley University, Qena 83523, Egypt; (W.M.); (T.A.); (E.A.A.)
| | - Reham Hassan
- Leibniz Research Centre for Working Environment and Human Factors at the Technical University Dortmund, 44139 Dortmund, Germany, (A.Z.); , (J.R.); (D.D.)
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, South Valley University, Qena 83523, Egypt
| | - Yasser A. Ahmed
- Department of Histology, Faculty of Veterinary Medicine, South Valley University, Qena 83523, Egypt;
| | - Tahany Abbas
- Histology Department, Faculty of Medicine, South Valley University, Qena 83523, Egypt; (W.M.); (T.A.); (E.A.A.)
| | - Eman A. Abdelrahim
- Histology Department, Faculty of Medicine, South Valley University, Qena 83523, Egypt; (W.M.); (T.A.); (E.A.A.)
| | - Kai Markus Schneider
- Department of Medicine III, University Hospital RWTH Aachen, Aachen University, 52074 Aachen, Germany; (K.M.S.); (M.-L.B.); (M.H.); (C.T.)
| | - Madlen Matz-Soja
- Faculty of Medicine, Rudolf-Schönheimer-Institute of Biochemistry, Leipzig University, 04103 Leipzig, Germany; (M.M.-S.); (R.G.)
| | - Jörg Reinders
- Leibniz Research Centre for Working Environment and Human Factors at the Technical University Dortmund, 44139 Dortmund, Germany, (A.Z.); , (J.R.); (D.D.)
| | - Rolf Gebhardt
- Faculty of Medicine, Rudolf-Schönheimer-Institute of Biochemistry, Leipzig University, 04103 Leipzig, Germany; (M.M.-S.); (R.G.)
| | - Marie-Luise Berres
- Department of Medicine III, University Hospital RWTH Aachen, Aachen University, 52074 Aachen, Germany; (K.M.S.); (M.-L.B.); (M.H.); (C.T.)
| | - Maximilian Hatting
- Department of Medicine III, University Hospital RWTH Aachen, Aachen University, 52074 Aachen, Germany; (K.M.S.); (M.-L.B.); (M.H.); (C.T.)
| | - Dirk Drasdo
- Leibniz Research Centre for Working Environment and Human Factors at the Technical University Dortmund, 44139 Dortmund, Germany, (A.Z.); , (J.R.); (D.D.)
- Modelling and Analysis for Medical and Biological Applications (MAMBA), Inria Paris & Sorbonne Université LJLL, 2 Rue Simone IFF, 75012 Paris, France
| | - Julio Saez-Rodriguez
- Faculty of Medicine, Institute of Computational Biomedicine, Heidelberg University, Bioquant—Im Neuenheimer Feld 267, 69120 Heidelberg, Germany; (C.H.H.); (J.S.-R.)
- Faculty of Medicine, Joint Research Centre for Computational Biomedicine (JRC-COMBINE), RWTH Aachen University, Pauwelsstrasse 19, 52074 Aachen, Germany
| | - Christian Trautwein
- Department of Medicine III, University Hospital RWTH Aachen, Aachen University, 52074 Aachen, Germany; (K.M.S.); (M.-L.B.); (M.H.); (C.T.)
| | - Jan G. Hengstler
- Leibniz Research Centre for Working Environment and Human Factors at the Technical University Dortmund, 44139 Dortmund, Germany, (A.Z.); , (J.R.); (D.D.)
| |
Collapse
|
19
|
Comparative transcriptome analysis of peripheral blood mononuclear cells in renal transplant recipients in everolimus- and tacrolimus-based immunosuppressive therapy. Eur J Pharmacol 2019; 859:172494. [PMID: 31238062 DOI: 10.1016/j.ejphar.2019.172494] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 06/21/2019] [Accepted: 06/21/2019] [Indexed: 12/19/2022]
Abstract
To better define the biological impact of immunosuppression on peripheral blood mononuclear cells (PBMC), we employed RNASeq analysis to compare the whole transcriptomic profile of a group of renal transplant recipients undergoing maintenance treatment with Everolimus (EVE) with those treated with Tacrolimus (TAC). Then, obtained results were validated by classical biomolecular methodologies. The statistical analysis allowed the identification of four genes discriminating the 2 study groups: Sushi Domain Containing 4 (SUSD4, P = 0.02), T Cell Leukemia/Lymphoma 1A (TCL1A, P = 0.02), adhesion G protein-coupled receptor E3 (ADGRE3, P = 0.01), Immunoglobulin Heavy Constant Gamma 3 (IGHG3, P = 0.03). All of them were significantly down-regulated in patients treated with EVE compared to TAC. The Area under Receiver Operating Characteristic (AUROC) of the final model based on these 4 genes was 73.1% demonstrating its good discriminative power. RT-PCR and ELISA validated transcriptomic results. Additionally, an in vitro model confirmed that EVE significantly down-regulates (P<0.001) TCL1A, SUSD4, ADGRE3 and IgHG3 in PBMCs as well as in T cells and monocytes isolated from healthy subjects. Taken together, our data, revealed, for the first time, a new four gene-based transcriptomic fingerprint down-regulated by EVE in PBMCs of renal transplant patients that could improve the available knowledge regarding some of the biological/cellular effects of the mTOR-Is (including their antineoplastic and immune-regulatory properties).
Collapse
|
20
|
Association between the pig genome and its gut microbiota composition. Sci Rep 2019; 9:8791. [PMID: 31217427 PMCID: PMC6584621 DOI: 10.1038/s41598-019-45066-6] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 05/30/2019] [Indexed: 12/24/2022] Open
Abstract
The gut microbiota has been evolving with its host along the time creating a symbiotic relationship. In this study, we assess the role of the host genome in the modulation of the microbiota composition in pigs. Gut microbiota compositions were estimated through sequencing the V3-V4 region of the 16S rRNA gene from rectal contents of 285 pigs. A total of 1,261 operational taxonomic units were obtained and grouped in 18 phyla and 101 genera. Firmicutes (45.36%) and Bacteroidetes (37.47%) were the two major phyla obtained, whereas at genus level Prevotella (7.03%) and Treponema (6.29%) were the most abundant. Pigs were also genotyped with a high-throughput method for 45,508 single nucleotide polymorphisms that covered the entire pig genome. Subsequently, genome-wide association studies were made among the genotypes of these pigs and their gut microbiota composition. A total of 52 single-nucleotide polymorphisms distributed in 17 regions along the pig genome were associated with the relative abundance of six genera; Akkermansia, CF231, Phascolarctobacterium, Prevotella, SMB53, and Streptococcus. Our results suggest 39 candidate genes that may be modulating the microbiota composition and manifest the association between host genome and gut microbiota in pigs.
Collapse
|
21
|
Novel potential inhibitors of complement system and their roles in complement regulation and beyond. Mol Immunol 2018; 102:73-83. [PMID: 30217334 DOI: 10.1016/j.molimm.2018.05.023] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 05/02/2018] [Accepted: 05/25/2018] [Indexed: 12/20/2022]
Abstract
The complement system resembles a double-edged sword since its activation can either benefit or harm the host. Thus, regulation of this system is of utmost importance and performed by several circulating and membrane-bound complement inhibitors. The pool of well-established regulators has recently been enriched with proteins that either share structural homology to known complement inhibitors such as Sushi domain-containing (SUSD) protein family and Human CUB and Sushi multiple domains (CSMD) families or extracellular matrix (ECM) macromolecules that interact with and modulate complement activity. In this review, we summarize the current knowledge about newly discovered complement inhibitors and discuss their implications in complement regulation, as well as in processes beyond complement regulation such cancer development. Understanding the behavior of these proteins will introduce new mechanisms of complement regulation and may provide new avenues in the development of novel therapies.
Collapse
|
22
|
Silawal S, Triebel J, Bertsch T, Schulze-Tanzil G. Osteoarthritis and the Complement Cascade. CLINICAL MEDICINE INSIGHTS. ARTHRITIS AND MUSCULOSKELETAL DISORDERS 2018; 11:1179544117751430. [PMID: 29434479 PMCID: PMC5805003 DOI: 10.1177/1179544117751430] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 12/07/2017] [Indexed: 12/11/2022]
Abstract
Accumulating evidence demonstrates that complement activation is involved in the pathogenesis of osteoarthritis (OA). However, the intimate complement regulation and cross talk with other signaling pathways in joint-associated tissues remain incompletely understood. Recent insights are summarized and discussed here, to put together a more comprehensive picture of complement involvement in OA pathogenesis. Complement is regulated by several catabolic and inflammatory mediators playing a key role in OA. It seems to be involved in many processes observed during OA development and progression, such as extracellular cartilage matrix (ECM) degradation, chondrocyte and synoviocyte inflammatory responses, cell lysis, synovitis, disbalanced bone remodeling, osteophyte formation, and stem cell recruitment, as well as cartilage angiogenesis. In reverse, complement can be activated by various ECM components and their cleavage products, which are released during OA-associated cartilage degradation. There are, however, some other cartilage ECM components that can inhibit complement, underlining the diverse effects of ECM on the complement activation. It is hypothesized that complement might also be directly activated by mechanical stress, thereby contributing to OA. The question arises whether keeping the complement activation in balance could represent a future therapeutic strategy in OA treatment and in the prevention of its progression.
Collapse
Affiliation(s)
- Sandeep Silawal
- Department of Anatomy, Paracelsus Medical University, Nuremberg, Germany
- Institute of Anatomy, Paracelsus Medical University, Salzburg, Germany
| | - Jakob Triebel
- Institute for Clinical Chemistry, Laboratory Medicine and Transfusion Medicine, Nuremberg General Hospital, Paracelsus Medical University, Nuremberg, Germany
| | - Thomas Bertsch
- Institute for Clinical Chemistry, Laboratory Medicine and Transfusion Medicine, Nuremberg General Hospital, Paracelsus Medical University, Nuremberg, Germany
| | - Gundula Schulze-Tanzil
- Department of Anatomy, Paracelsus Medical University, Nuremberg, Germany
- Institute of Anatomy, Paracelsus Medical University, Salzburg, Germany
| |
Collapse
|
23
|
Abstract
The complement system is an arm of innate immunity that aids in the removal of pathogens and dying cells. Due to its harmful, pro-inflammatory potential, complement is controlled by several soluble and membrane-bound inhibitors. This family of complement regulators has been recently extended by the discovery of several new members, and it is becoming apparent that these proteins harbour additional functions. In this review, the current state of knowledge of the physiological functions of four complement regulators will be described: cartilage oligomeric matrix protein (COMP), CUB and sushi multiple domains 1 (CSMD1), sushi domain-containing protein 4 (SUSD4) and CD59. Complement activation is involved in both the development of and defence against cancer. COMP expression is pro-oncogenic, whereas CSMD1 and SUSD4 act as tumour suppressors. These effects may be related in part to the complex influence of complement on cancer but also depend on unrelated functions such as the protection of cells from endoplasmic reticulum stress conveyed by intracellular COMP. CD59 is the main inhibitor of the membrane attack complex, and its deficiency leads to complement attack on erythrocytes and severe haemolytic anaemia, which is now amenable to treatment with an inhibitor of C5 cleavage. Unexpectedly, the intracellular pool of CD59 is crucial for insulin secretion from pancreatic β-cells. This finding is one of several relating to the intracellular functions of complement proteins, which until recently were only considered to be present in the extracellular space. Understanding the alternative functions of complement inhibitors may unravel unexpected links between complement and other physiological systems, but is also important for better design of therapeutic complement inhibition.
Collapse
Affiliation(s)
- A M Blom
- Division of Medical Protein Chemistry, Department of Translational Medicine, Lund University, Malmö, Sweden
| |
Collapse
|
24
|
RNA-Seq reveals common and unique PXR- and CAR-target gene signatures in the mouse liver transcriptome. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2016; 1859:1198-1217. [PMID: 27113289 DOI: 10.1016/j.bbagrm.2016.04.010] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 04/19/2016] [Accepted: 04/19/2016] [Indexed: 12/14/2022]
Abstract
The pregnane X receptor (PXR) and constitutive androstane receptor (CAR) are well-known xenobiotic-sensing nuclear receptors with overlapping functions. However, there lacks a quantitative characterization to distinguish between the PXR and CAR target genes and signaling pathways in the liver. The present study performed a transcriptomic comparison of the PXR- and CAR-targets using RNA-Seq in livers of adult wild-type mice that were treated with the prototypical PXR ligand PCN (200mg/kg, i.p. once daily for 4days in corn oil) or the prototypical CAR ligand TCPOBOP (3mg/kg, i.p., once daily for 4days in corn oil). At the given doses, TCPOBOP differentially regulated many more genes (2125) than PCN (212), and 147 of the same genes were differentially regulated by both chemicals. As expected, the top pathways differentially regulated by both PCN and TCPOBOP were involved in xenobiotic metabolism, and they also up-regulated genes involved in retinoid metabolism, but down-regulated genes involved in inflammation and iron homeostasis. Regarding unique pathways, PXR activation appeared to overlap with the aryl hydrocarbon receptor signaling, whereas CAR activation appeared to overlap with the farnesoid X receptor signaling, acute-phase response, and mitochondrial dysfunction. The mRNAs of differentially regulated drug-processing genes (DPGs) partitioned into three patterns, namely TCPOBOP-induced, PCN-induced, as well as TCPOBOP-suppressed gene clusters. The cumulative mRNAs of the differentially regulated DPGs, phase-I and -II enzymes, as well as efflux transporters were all up-regulated by both PCN and TCPOBOPOP, whereas the cumulative mRNAs of the uptake transporters were down-regulated only by TCPOBOP. The absolute mRNA abundance in control and receptor-activated conditions was examined in each DPG category to predict the contribution of specific DPG genes in the PXR/CAR-mediated pharmacokinetic responses. The preferable differential regulation by TCPOBOP in the entire hepatic transcriptome correlated with a marked change in the expression of many DNA and histone epigenetic modifiers. In conclusion, the present study has revealed known and novel, as well as common and unique targets of PXR and CAR in mouse liver following pharmacological activation using their prototypical ligands. Results from this study will further support the role of these receptors in regulating the homeostasis of xenobiotic and intermediary metabolism in the liver, and aid in distinguishing between PXR and CAR signaling at various physiological and pathophysiological conditions. This article is part of a Special Issue entitled: Xenobiotic nuclear receptors: New Tricks for An Old Dog, edited by Dr. Wen Xie.
Collapse
|
25
|
Abstract
OBJECTIVE To identify genetic variants associated with the severity of postburn hypertrophic scarring (HTS) using a genome-wide approach. BACKGROUND Risk of severe postburn HTS is known to depend on race, but the genetic determinants of HTS are unknown. METHODS We conducted a genome-wide association study (GWAS) in a prospective cohort of adults admitted with deep-partial-thickness burns from 2007 through 2014. Scar severity was assessed over time using the Vancouver Scar Scale (VSS), and DNA was genotyped with a >500,000-marker array. We performed association testing of single-nucleotide polymorphisms (SNPs) with minor allele frequency (MAF) >0.01 using linear regression of VSS height score on genotype adjusted for patient and injury characteristics as well as population genetic structure. Array-wide significance was based on Bonferroni correction for multiple testing. RESULTS Of 538 patients (median age 40 years, median burn size 6.0% of body surface area), 71% were men and 76% were White. The mean VSS height score was 1.2 (range: 0-3). Of 289,639 SNPs tested, a variant in the CUB and Sushi multiple domains 1 (CSMD1) gene (rs11136645; MAF = 0.49), was significantly associated with decreased scar height (regression coefficient = -0.23, P = 7.9 × 10). CONCLUSIONS In the first published GWAS of HTS, we report that a common intronic variant in the CSMD1 gene is associated with reduced severity of postburn HTS. If this association is confirmed in an independent cohort, investigating the potential role of CSMD1 in wound healing may elucidate HTS pathophysiology.
Collapse
|
26
|
Englund E, Reitsma B, King BC, Escudero-Esparza A, Owen S, Orimo A, Okroj M, Anagnostaki L, Jiang WG, Jirström K, Blom AM. The human complement inhibitor Sushi Domain-Containing Protein 4 (SUSD4) expression in tumor cells and infiltrating T cells is associated with better prognosis of breast cancer patients. BMC Cancer 2015; 15:737. [PMID: 26480818 PMCID: PMC4615997 DOI: 10.1186/s12885-015-1734-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 10/08/2015] [Indexed: 01/22/2023] Open
Abstract
Background The human Sushi Domain-Containing Protein 4 (SUSD4) was recently shown to function as a novel inhibitor of the complement system, but its role in tumor progression is unknown. Methods Using immunohistochemistry and quantitative PCR, we investigated SUSD4 expression in breast cancer tissue samples from two cohorts. The effect of SUSD4 expression on cell migration and invasion was studied in vitro using two human breast cancer cell lines overexpressing SUSD4. Results Tissue stainings revealed that both tumor cells and tumor-infiltrating cells expressed SUSD4. The highest SUSD4 expression was detected in differentiated tumors with decreased rate of metastasis, and SUSD4 expression was associated with improved survival of the patients. Moreover, forced SUSD4 expression in human breast cancer cells attenuated their migratory and invasive traits in culture. SUSD4 expression also inhibited colony formation of human breast cancer cells cultured on carcinoma-associated fibroblasts. Furthermore, large numbers of SUSD4-expressing T cells in the tumor stroma associated with better overall survival of the breast cancer patients. Conclusion Our findings indicate that SUSD4 expression in both breast cancer cells and T cells infiltrating the tumor-associated stroma is useful to predict better prognosis of breast cancer patients.
Collapse
Affiliation(s)
- Emelie Englund
- Department of Translational Medicine, Division of Medical Protein Chemistry, Lund University, Inga Maria Nilssons gata 53, 20502, Malmö, Sweden.
| | - Bart Reitsma
- Department of Translational Medicine, Division of Medical Protein Chemistry, Lund University, Inga Maria Nilssons gata 53, 20502, Malmö, Sweden.
| | - Ben C King
- Department of Translational Medicine, Division of Medical Protein Chemistry, Lund University, Inga Maria Nilssons gata 53, 20502, Malmö, Sweden.
| | - Astrid Escudero-Esparza
- Department of Translational Medicine, Division of Medical Protein Chemistry, Lund University, Inga Maria Nilssons gata 53, 20502, Malmö, Sweden.
| | - Sioned Owen
- Cardiff's China Medical Research Collaborative (CCMRC), Cardiff University School of Medicine, Cardiff University, Cardiff, UK.
| | - Akira Orimo
- Department of Pathology and Oncology, Juntendo University School of Medicine, Tokyo, Japan.
| | - Marcin Okroj
- Department of Translational Medicine, Division of Medical Protein Chemistry, Lund University, Inga Maria Nilssons gata 53, 20502, Malmö, Sweden. .,Department of Medical Biotechnology, Intercollegiate Faculty of Biotechnology UG‑MUG, Medical University of Gdańsk, 80210, Gdańsk, Poland.
| | - Lola Anagnostaki
- Department of Clinical Pathology, Skåne University Hospital, Malmö, Sweden.
| | - Wen G Jiang
- Cardiff's China Medical Research Collaborative (CCMRC), Cardiff University School of Medicine, Cardiff University, Cardiff, UK.
| | - Karin Jirström
- Department of Clinical Sciences Lund, Division of Oncology and Pathology, Lund University, Lund, Sweden.
| | - Anna M Blom
- Department of Translational Medicine, Division of Medical Protein Chemistry, Lund University, Inga Maria Nilssons gata 53, 20502, Malmö, Sweden.
| |
Collapse
|
27
|
Abstract
Although new activation and regulatory mechanisms are still being identified, the basic architecture of the complement system has been known for decades. Two major roles of complement are to control certain bacterial infections and to promote clearance of apoptotic cells. In addition, although inappropriate complement activation has long been proposed to cause tissue damage in human inflammatory and autoimmune diseases, whether this is indeed true has been uncertain. However, recent studies in humans, especially those using newly available biological therapeutics, have now clearly demonstrated the pathophysiologic importance of the complement system in several rare diseases. Beyond these conditions, recent genetic studies have strongly supported an injurious role for complement in a wide array of human inflammatory, degenerative, and autoimmune diseases. This review includes an overview of complement activation, regulatory, and effector mechanisms. It then focuses on new understandings gained from genetic studies, ex vivo analyses, therapeutic trials, and animal models as well as on new research opportunities.
Collapse
Affiliation(s)
- V Michael Holers
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado 80045;
| |
Collapse
|
28
|
Shen Y, Yang L, Li R. What does complement do in Alzheimer's disease? Old molecules with new insights. Transl Neurodegener 2013; 2:21. [PMID: 24119446 PMCID: PMC3853043 DOI: 10.1186/2047-9158-2-21] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Accepted: 10/01/2013] [Indexed: 11/25/2022] Open
Abstract
Increasing evidence suggests that inflammatory and immune components in brain are important in Alzheimer's disease (AD) and anti-inflammatory and immunotherapeutic approaches may be amenable to AD treatment. It is known that complement activation occurs in the brain of patients with AD, and contributes to a local inflammatory state development which is correlated with cognitive impairment. In addition to the complement's critical role in the innate immune system recognizing and killing, or targeting for destruction, complement proteins can also interact with cell surface receptors to promote a local inflammatory response and contributes to the protection and healing of the host. On the other hand, complement activation also causes inflammation and cell damage as an essential immune function to eliminate cell debris and potentially toxic protein aggregates. It is the balance of these seemingly competing events that influences the ultimate state of neuronal function. Our mini review will be focusing on the unique molecular interactions happening in the AD development, the functional outcomes of those interactions, as well as the contribution of each element to AD.
Collapse
Affiliation(s)
- Yong Shen
- Center for Advanced Therapeutic Strategies for Brain Disorders, Roskamp Institute, 2040 Whitfield Avenue, Sarasota, USA
- Department of Neurology, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Libang Yang
- Department of Pediatrics, School of Medicine, University of Minnesota, Minneapolis, MN 55455, USA
| | - Rena Li
- Center for Hormones Advanced Science and Education, Roskamp Institute, 2040 Whitfield Avenue, Sarasota, Florida, USA
| |
Collapse
|