1
|
Gilbert FB, Martins RP, Rainard P. Expression of FcμR by bovine mononuclear blood leukocytes. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2024; 162:105304. [PMID: 39674305 DOI: 10.1016/j.dci.2024.105304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 11/20/2024] [Accepted: 12/11/2024] [Indexed: 12/16/2024]
Abstract
The receptor for IgM has been identified a few years ago, but its expression by bovine mononuclear cells has not yet been studied. We used rabbit antibodies against bovine FcμR to begin to fill this gap. Anti-FcμR antibodies bound to B lymphocytes and monocytes, although less than to neutrophils. Nonclassical and intermediate monocytes (CD172apos CD16pos) bound nonspecifically to rabbit antibodies, complicating analysis, but they bound more anti-FcμR antibodies than control antibodies, indicating that they also express the FcμR. They also express more C5a receptors than classical monocytes. Anti-FcμR antibodies did not bind to CD3pos αβT lymphocytes (both CD4pos and CD8pos) and γδT cells. At low temperature but not at physiological temperature, purified bovine IgM bound to all monocytes and strongly to all B cells, but hardly to CD3pos T cells. Monocytes and B cells bound human IgA, but IgA did not compete, whereas unlabeled bovine IgM competed for binding of labeled IgM. This supports the role of the FcμR, and not the FαμR, in IgM binding. Finally, we showed that monocytes were able to ingest bacteria opsonized with serum deprived of IgG, indicating their ability to perform IgM-dependent phagocytosis. In conclusion, surface expression of FcμR by unstimulated blood leukocytes was demonstrated on B cells and monocytes, but not on T cells.
Collapse
|
2
|
Li S, Ge F, Chen L, Liu Y, Chen Y, Ma Y. Genome-wide association analysis of body conformation traits in Chinese Holstein Cattle. BMC Genomics 2024; 25:1174. [PMID: 39627684 PMCID: PMC11616231 DOI: 10.1186/s12864-024-11090-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 11/26/2024] [Indexed: 12/06/2024] Open
Abstract
BACKGROUND The body conformation traits of dairy cattle are closely related to their production performance and health. The present study aimed to identify gene variants associated with body conformation traits in Chinese Holstein cattle and provide marker loci for genomic selection in dairy cattle breeding. These findings could offer robust theoretical support for optimizing the health of dairy cattle and enhancing their production performance. RESULTS This study involved 586 Chinese Holstein cattle and used the predicted transmitting abilities (PTAs) of 17 body conformation traits evaluated by the Council on Dairy Cattle Breeding in the USA as phenotypic values. These traits were categorized into body size traits, rump traits, feet/legs traits, udder traits, and dairy characteristic traits. On the basis of the genomic profiling results from the Genomic Profiler Bovine 100 K SNP chip, genotype data were quality controlled via PLINK software, and 586 individuals and 80,713 SNPs were retained for further analysis. Genome-wide association studies (GWASs) were conducted via GEMMA software, which employs both univariate linear mixed models (LMMs) and multivariate linear mixed models (mvLMMs). The Bonferroni method was used to determine the significance threshold, identifying gene variants significantly associated with body conformation traits in Chinese Holstein cattle. The single-trait GWAS identified 24 SNPs significantly associated with body conformation traits (P < 0.01), with annotation leading to the identification of 21 candidate genes. The multi-trait GWAS identified 54 SNPs, which were annotated to 57 candidate genes, including 39 new SNPs not identified in the single-trait GWAS. Additionally, 14 SNPs in the 86.84-87.41 Mb region of chromosome 6 were significantly associated with multiple traits, such as body size, udder, and dairy characteristics. Four genes-SLC4A4, GC, NPFFR2, and ADAMTS3-were annotated in this region. CONCLUSIONS A total of 63 SNPs were identified as significantly associated with 17 body conformation traits in Chinese Holstein cattle through both single-trait and multi-trait GWAS analyses. Sixty-six candidate genes were annotated, with 12 genes identified by both methods, such as SLC4A4, GC, NPFFR2, and ADAMTS3, which are involved in pathways such as growth hormone synthesis and secretion, sphingolipid signaling, and dopaminergic synapse pathways. These findings provide potential genetic marker information related to body conformation traits for the breeding of Chinese Holstein cattle.
Collapse
Affiliation(s)
- Shuangshuang Li
- Tianjin Key Laboratory of Animal Molecular Breeding and Biotechnology, Tianjin Engineering Research Center of Animal Healthy Farming, Institute of Animal Science and Veterinary, Tianjin Academy of Agricultural Sciences, Tianjin, 300381, China
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, 300392, China
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences (CAAS), Beijing, 100193, China
| | - Fei Ge
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences (CAAS), Beijing, 100193, China
| | - Lili Chen
- Tianjin Key Laboratory of Animal Molecular Breeding and Biotechnology, Tianjin Engineering Research Center of Animal Healthy Farming, Institute of Animal Science and Veterinary, Tianjin Academy of Agricultural Sciences, Tianjin, 300381, China
| | - Yuxin Liu
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences (CAAS), Beijing, 100193, China
| | - Yan Chen
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences (CAAS), Beijing, 100193, China.
| | - Yi Ma
- Tianjin Key Laboratory of Animal Molecular Breeding and Biotechnology, Tianjin Engineering Research Center of Animal Healthy Farming, Institute of Animal Science and Veterinary, Tianjin Academy of Agricultural Sciences, Tianjin, 300381, China.
| |
Collapse
|
3
|
Talker SC, Hope JC, Summerfield A. Phenotype of bovine mononuclear phagocytes- An update. Vet Immunol Immunopathol 2024; 277:110836. [PMID: 39368394 DOI: 10.1016/j.vetimm.2024.110836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 09/22/2024] [Accepted: 09/23/2024] [Indexed: 10/07/2024]
Abstract
Studying mononuclear phagocytes by flow cytometry is challenging due to their phenotypic similarities and the high plasticity of monocytic cells. Despite these challenges, significant progress has been made in cattle research through multicolor flow cytometry, transcriptomics of sorted subsets, and single-cell RNA-sequencing. Here, we provide an overview of established and proposed phenotypic classifications in the bovine mononuclear phagocyte system and discuss the challenges of marker discovery.
Collapse
Affiliation(s)
- S C Talker
- Institute of Virology and Immunology, Bern, Switzerland; Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland.
| | - J C Hope
- The Roslin Institute, University of Edinburgh, Easter Bush, EH25 9RG, UK
| | - A Summerfield
- Institute of Virology and Immunology, Bern, Switzerland; Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| |
Collapse
|
4
|
Krogstad KC, Fehn JF, Mamedova LK, Bernard MP, Bradford BJ. Effects of rumen-protected niacin on inflammatory response to repeated intramammary lipopolysaccharide challenges. J Dairy Sci 2024:S0022-0302(24)00927-5. [PMID: 38876216 DOI: 10.3168/jds.2024-24974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 05/15/2024] [Indexed: 06/16/2024]
Abstract
Nutritional strategies that improve an animal's resilience to various challenges may improve animal health and welfare. One such nutrient is niacin which has reduced inflammation in mice, humans, and swine; however, niacin's anti-inflammatory effects have not been investigated in cattle. Our objective was to determine whether rumen-protected niacin (RPN) alters lactating dairy cows' inflammatory response to intramammary lipopolysaccharide (LPS) challenges, whether RPN resulted in any carry-over effects, and whether repeated LPS challenges result in signs of immune tolerance or innate immune training. Twenty healthy, late-lactation Holstein cows (232 ± 65 d in milk; 39 ± 5.8 kg/d of milk) were enrolled in a randomized complete block experiment which lasted 70 d. Cows received 26 g/d of RPN or no top-dress (CON) for the first 42 d of the experiment. During the final milking of d 27 and 55, cows were challenged in their rear-right mammary gland (RR) with 100 µg of LPS suspended in 5 mL of phosphate buffered saline. Milk yield, milk conductivity, and feed intake were measured daily. Milk composition was measured on d 14, 23, 24, 30, 37, 45, and 52. Blood samples were collected at 0, 8, 12, 24, 48, 72, 96, and 120 h after each LPS challenge, whereas RR quarter milk samples were collected at 0, 8, 16, 24, 48, 72, 96, 120, 144, and 168 h after each LPS challenge. Body temperature was measured continuously during each challenge with an intravaginal thermometer. Linear mixed models with repeated measures were used to analyze the results. Before LPS challenge, RPN did not affect feed intake or milk production, but it reduced SCS (1.24 ± 0.41 vs. 0.05 ± 0.45). After challenge, RPN did not affect feed intake, milk production, milk composition, SCS, body temperature, plasma glucose, or plasma insulin concentrations. Our results suggest RPN reduced peak plasma haptoglobin and lipopolysaccharide binding protein (LBP) during the 1st LPS challenge. Plasma haptoglobin tended to be less after the 2nd challenge for cows previously supplemented RPN while LBP was similar for each treatment group after the 2nd challenge. The 2nd LPS challenge resulted in decreased plasma haptoglobin compared with the 1st LPS challenge, suggestive of tolerance but it also induced a greater peak SCS than the 1st LPS challenge. Our results suggest that repeated LPS challenges promote a systemic tolerance but heightened local response to LPS-induced mastitis. Feeding RPN reduced SCS before challenge and reduced plasma acute phase proteins after challenge suggesting that RPN may reduce systemic inflammation without altering the local inflammatory responses.
Collapse
Affiliation(s)
- K C Krogstad
- Department of Animal Science, Michigan State University, East Lansing 48824; Department of Animal Science, The Ohio State University, Wooster, OH 44691 USA.
| | - J F Fehn
- Department of Animal Science, Michigan State University, East Lansing 48824
| | - L K Mamedova
- Department of Animal Science, Michigan State University, East Lansing 48824
| | - M P Bernard
- Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, East Lansing, Michigan, 48824 USA; Department of Pharmacology & Toxicology, Michigan State University, East Lansing, MI, 48824 USA
| | - B J Bradford
- Department of Animal Science, Michigan State University, East Lansing 48824
| |
Collapse
|
5
|
Danev N, Li G, Duan J(E, Van de Walle GR. Comparative transcriptomic analysis of bovine mesenchymal stromal cells reveals tissue-source and species-specific differences. iScience 2024; 27:108886. [PMID: 38318381 PMCID: PMC10838956 DOI: 10.1016/j.isci.2024.108886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 11/27/2023] [Accepted: 01/08/2024] [Indexed: 02/07/2024] Open
Abstract
Mesenchymal stromal cells (MSCs) have the potential to be used as therapeutics, but their efficacy varies due to cellular heterogeneity, which is not fully understood. After characterizing donor-matched bovine MSC from adipose tissue (AT), bone marrow (BM), and peripheral blood (PB), we performed single-cell RNA sequencing (scRNA-seq) to evaluate overarching similarities and differences across these three tissue-derived MSCs. Next, the transcriptomic profiles of the bovine MSCs were compared to those of equine MSCs, derived from the same tissue sources and previously published by our group, and revealed species-specific differences. Finally, the transcriptomic profile from bovine BM-MSCs was compared to mouse and human BM-MSCs and demonstrated that bovine BM-MSCs share more common functionally relevant gene expression profiles with human BM-MSCs than compared to murine BM-MSCs. Collectively, this study presents the cow as a potential non-traditional animal model for translational MSC studies based on transcriptomic profiles similar to human MSCs.
Collapse
Affiliation(s)
- Nikola Danev
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Guangsheng Li
- Department of Animal Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Jingyue (Ellie) Duan
- Department of Animal Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Gerlinde R. Van de Walle
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
6
|
Démoulins T, Yimthin T, Lindtke D, Eggerschwiler L, Siegenthaler R, Labroussaa F, Jores J. Temperature impacts the bovine ex vivo immune response towards Mycoplasmopsis bovis. Vet Res 2024; 55:18. [PMID: 38351086 PMCID: PMC10863263 DOI: 10.1186/s13567-024-01272-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 01/13/2024] [Indexed: 02/16/2024] Open
Abstract
Although cattle are the mammalian species with most global biomass associated with a huge impact on our planet, their immune system remains poorly understood. Notably, the bovine immune system has peculiarities such as an overrepresentation of γδ T cells that requires particular attention, specifically in an infectious context. In line of 3R principles, we developed an ex vivo platform to dissect host-pathogen interactions. The experimental design was based on two independent complementary readouts: firstly, a novel 12-14 color multiparameter flow cytometry assay measuring maturation (modulation of cell surface marker expression) and activation (intracellular cytokine detection) of monocytes, conventional and plasmacytoid dendritic cells, natural killer cells, γδ T cells, B and T cells; secondly, a multiplex immunoassay monitoring bovine chemokine and cytokine secretion levels. The experiments were conducted on fresh primary bovine blood cells exposed to Mycoplasmopsis bovis (M. bovis), a major bovine respiratory pathogen. Besides reaffirming the tight cooperation of the different primary blood cells, we also identified novel key players such as strong IFN-γ secreting NK cells, whose role was so far largely overlooked. Additionally, we compared the host-pathogen interactions at different temperatures, including commonly used 37 °C, ruminant body temperature (38-38.5 °C) and fever (≥ 39.5 °C). Strikingly, working under ruminant physiological temperature influenced the capacity of most immune cell subsets to respond to M. bovis compared to 37 °C. Under fever-like temperature conditions the immune response was impaired compared to physiological temperature. Our experimental approach, phenotypically delineating the bovine immune system provided a thorough vision of the immune response towards M. bovis and the influence of temperature towards that immune response.
Collapse
Affiliation(s)
- Thomas Démoulins
- Institute of Veterinary Bacteriology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, 3001, Bern, Switzerland.
| | - Thatcha Yimthin
- Institute of Veterinary Bacteriology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, 3001, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Dorothea Lindtke
- Animal GenoPhenomics Group, Agroscope, 1725, Posieux, Switzerland
| | | | | | - Fabien Labroussaa
- Institute of Veterinary Bacteriology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, 3001, Bern, Switzerland
- Multidisciplinary Center for Infectious Diseases (MCID), University of Bern, 3001, Bern, Switzerland
| | - Joerg Jores
- Institute of Veterinary Bacteriology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, 3001, Bern, Switzerland
- Multidisciplinary Center for Infectious Diseases (MCID), University of Bern, 3001, Bern, Switzerland
| |
Collapse
|
7
|
Louloudes-Lázaro A, Rojas JM, García-García I, Rodríguez-Martín D, Morel E, Martín V, Sevilla N. Comprehensive immune profiling reveals that Orbivirus infection activates immune checkpoints during acute T cell immunosuppression. Front Immunol 2023; 14:1255803. [PMID: 37920474 PMCID: PMC10619675 DOI: 10.3389/fimmu.2023.1255803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Accepted: 10/03/2023] [Indexed: 11/04/2023] Open
Abstract
Bluetongue virus (BTV) is an arbovirus transmitted by the bite of infected Culicoides midges that affects domestic and wild ruminants producing great economic losses. The infection induces an IFN response, followed by an adaptive immune response that is essential in disease clearance. BTV can nonetheless impair IFN and humoral responses. The main goal of this study was to gain a more detailed understanding of BTV pathogenesis and its effects on immune cell populations. To this end, we combined flow cytometry and transcriptomic analyses of several immune cells at different times post-infection (pi). Four sheep were infected with BTV serotype 8 and blood samples collected at days 0, 3, 7 and 15pi to perform transcriptomic analysis of B-cell marker+, CD4+, CD8+, and CD14+ sorted peripheral mononuclear cells. The maximum number of differentially expressed genes occurred at day 7pi, which coincided with the peak of infection. KEGG pathway enrichment analysis indicated that genes belonging to virus sensing and immune response initiation pathways were enriched at day 3 and 7 pi in all 4 cell population analyzed. Transcriptomic analysis also showed that at day 7pi T cell exhaustion pathway was enriched in CD4+ cells, while CD8+ cells downregulated immune response initiation pathways. T cell functional studies demonstrated that BTV produced an acute inhibition of CD4+ and CD8+ T cell activation at the peak of replication. This coincided with PD-L1 upregulation on the surface of CD4+ and CD8+ T cells as well as monocytes. Taken together, these data indicate that BTV could exploit the PD1/PD-L1 immune checkpoint to impair T cell responses. These findings identify several mechanisms in the interaction between host and BTV, which could help develop better tools to combat the disease.
Collapse
Affiliation(s)
- Andrés Louloudes-Lázaro
- Centro de Investigación en Sanidad Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Consejo Superior de Investigaciones Científicas (CISA-INIA-CSIC), Madrid, Spain
| | - José M. Rojas
- Centro de Investigación en Sanidad Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Consejo Superior de Investigaciones Científicas (CISA-INIA-CSIC), Madrid, Spain
| | - Isabel García-García
- Departamento de Genética, Fisiología y Microbiología, Unidad de Genética, Facultad de Ciencias Biológicas, Universidad Complutense de Madrid (UCM), Madrid, Spain
| | - Daniel Rodríguez-Martín
- Centro de Investigación en Sanidad Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Consejo Superior de Investigaciones Científicas (CISA-INIA-CSIC), Madrid, Spain
| | - Esther Morel
- Centro de Investigación en Sanidad Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Consejo Superior de Investigaciones Científicas (CISA-INIA-CSIC), Madrid, Spain
| | - Verónica Martín
- Centro de Investigación en Sanidad Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Consejo Superior de Investigaciones Científicas (CISA-INIA-CSIC), Madrid, Spain
| | - Noemí Sevilla
- Centro de Investigación en Sanidad Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Consejo Superior de Investigaciones Científicas (CISA-INIA-CSIC), Madrid, Spain
| |
Collapse
|
8
|
Worku D, Hussen J, De Matteis G, Schusser B, Alhussien MN. Candidate genes associated with heat stress and breeding strategies to relieve its effects in dairy cattle: a deeper insight into the genetic architecture and immune response to heat stress. Front Vet Sci 2023; 10:1151241. [PMID: 37771947 PMCID: PMC10527375 DOI: 10.3389/fvets.2023.1151241] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 08/31/2023] [Indexed: 09/30/2023] Open
Abstract
The need for food products of animal origin is increasing worldwide. Satisfying these needs in a way that has minimal impact on the environment requires cutting-edge technologies and techniques to enhance the genetic quality of cattle. Heat stress (HS), in particular, is affecting dairy cattle with increasing frequency and severity. As future climatic challenges become more evident, identifying dairy cows that are more tolerant to HS will be important for breeding dairy herds that are better adapted to future environmental conditions and for supporting the sustainability of dairy farming. While research into the genetics of HS in the context of the effect of global warming on dairy cattle is gaining momentum, the specific genomic regions involved in heat tolerance are still not well documented. Advances in omics information, QTL mapping, transcriptome profiling and genome-wide association studies (GWAS) have identified genomic regions and variants associated with tolerance to HS. Such studies could provide deeper insights into the genetic basis for response to HS and make an important contribution to future breeding for heat tolerance, which will help to offset the adverse effects of HS in dairy cattle. Overall, there is a great interest in identifying candidate genes and the proportion of genetic variation associated with heat tolerance in dairy cattle, and this area of research is currently very active worldwide. This review provides comprehensive information pertaining to some of the notable recent studies on the genetic architecture of HS in dairy cattle, with particular emphasis on the identified candidate genes associated with heat tolerance in dairy cattle. Since effective breeding programs require optimal knowledge of the impaired immunity and associated health complications caused by HS, the underlying mechanisms by which HS modulates the immune response and renders animals susceptible to various health disorders are explained. In addition, future breeding strategies to relieve HS in dairy cattle and improve their welfare while maintaining milk production are discussed.
Collapse
Affiliation(s)
- Destaw Worku
- Department of Animal Science, College of Agriculture, Food and Climate Sciences, Injibara University, Injibara, Ethiopia
| | - Jamal Hussen
- Department of Microbiology, College of Veterinary Medicine, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Giovanna De Matteis
- Council for Agricultural Research and Economics, CREA Research Centre for Animal Production and Aquaculture, Monterotondo, Rome, Italy
| | - Benjamin Schusser
- Reproductive Biotechnology, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Mohanned Naif Alhussien
- Reproductive Biotechnology, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
| |
Collapse
|
9
|
Grandoni F, Hussen J, Signorelli F, Napolitano F, Scatà MC, De Donato I, Cappelli G, Galiero G, Grassi C, De Carlo E, Petrini S, De Matteis G, Martucciello A. Evaluation of Hematological Profiles and Monocyte Subpopulations in Water Buffalo Calves after Immunization with Two Different IBR Marker Vaccines and Subsequent Infection with Bubaline alphaherpesvirus-1. Vaccines (Basel) 2023; 11:1405. [PMID: 37766082 PMCID: PMC10537172 DOI: 10.3390/vaccines11091405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/16/2023] [Accepted: 08/18/2023] [Indexed: 09/29/2023] Open
Abstract
Bubaline alphaherpesvirus-1 (BuAHV-1) and Bovine alphaherpesvirus-1 (BoAHV-1) are respiratory viruses that can cause an infection known as "Infectious Bovine Rhinotracheitis" (IBR) in both water buffalo and bovine species. As the main disease control strategy, vaccination can protect animals from clinical disease through the development of specific humoral and cell-mediated immune responses. In the present study, the time-related circulatory kinetics of hematological profile and bubaline monocyte subsets have been investigated in vaccinated buffalo calves after challenge infections with BuAHV-1. Thirteen buffalo calves were selected and grouped into the VAX-1 group, which received an IBR-live-attenuated gE-/tk-deleted marker vaccine; the VAX-2 group, which received an IBR-inactivated gE-deleted marker vaccine; the CNT group, which remained an unvaccinated control. Fifty-five days after the first vaccination, the animals were infected with 5 × 105.00 TCID50/mL of wild-type BuAHV-1 strain via the intranasal route. Whole blood samples were collected at 0, 2, 4, 7, 10, 15, 30, and 63 days post-challenge (PCDs) for the analysis of hematological profiles and the enumeration of monocyte subsets via flow cytometry. The analysis of leukocyte compositions revealed that neutrophils were the main leukocyte population, with a relative increase during the acute infection. On the other hand, a general decrease in the proportion of lymphocytes was observed early in the post-infection, both for the VAX-1 and VAX-2 groups, while in the CNT group, the decrease was observed later at +30 and +63 PCDs. An overall infection-induced increase in blood total monocytes was observed in all groups. The rise was especially marked in the animals vaccinated with an IBR-live-attenuated gE-/tK-deleted marker vaccine (VAX-1 group). A multicolor flow cytometry panel was used to identify the bubaline monocyte subpopulations (classical = cM; intermediate = intM; and non-classical = ncM) and to investigate their variations during BuAHV-1 infection. Our results showed an early increase in cMs followed by a second wave of intMs. This increase was observed mainly after stimulation with live-attenuated viruses in the VAX-1 group compared with the animals vaccinated with the inactivated vaccine or the non-vaccinated animal group. In summary, the present study characterized, for the first time, the hematological profile and distribution of blood monocyte subsets in vaccinated and non-vaccinated water buffalo in response to experimental infection with BuAHV-1. Although not experimentally proven, our results support the hypothesis of a linear developmental relationship between monocyte subsets.
Collapse
Affiliation(s)
- Francesco Grandoni
- Research Centre for Animal Production and Aquaculture, Consiglio per la Ricerca in Agricoltura e l’Analisi dell’Economia Agraria (CREA), 00015 Monterotondo, Italy
| | - Jamal Hussen
- Department of Microbiology, College of Veterinary Medicine, King Faisal University, Al-Ahsa 36362, Saudi Arabia
| | - Federica Signorelli
- Research Centre for Animal Production and Aquaculture, Consiglio per la Ricerca in Agricoltura e l’Analisi dell’Economia Agraria (CREA), 00015 Monterotondo, Italy
| | - Francesco Napolitano
- Research Centre for Animal Production and Aquaculture, Consiglio per la Ricerca in Agricoltura e l’Analisi dell’Economia Agraria (CREA), 00015 Monterotondo, Italy
| | - Maria Carmela Scatà
- Research Centre for Animal Production and Aquaculture, Consiglio per la Ricerca in Agricoltura e l’Analisi dell’Economia Agraria (CREA), 00015 Monterotondo, Italy
| | - Immacolata De Donato
- National Reference Centre for Hygiene and Technologies of Water Buffalo Farming and Productions, Istituto Zooprofilattico Sperimentale del Mezzogiorno, 84132 Salerno, Italy
| | - Giovanna Cappelli
- National Reference Centre for Hygiene and Technologies of Water Buffalo Farming and Productions, Istituto Zooprofilattico Sperimentale del Mezzogiorno, 84132 Salerno, Italy
| | - Giorgio Galiero
- National Reference Centre for Hygiene and Technologies of Water Buffalo Farming and Productions, Istituto Zooprofilattico Sperimentale del Mezzogiorno, 84132 Salerno, Italy
| | - Carlo Grassi
- National Reference Centre for Hygiene and Technologies of Water Buffalo Farming and Productions, Istituto Zooprofilattico Sperimentale del Mezzogiorno, 84132 Salerno, Italy
| | - Esterina De Carlo
- National Reference Centre for Hygiene and Technologies of Water Buffalo Farming and Productions, Istituto Zooprofilattico Sperimentale del Mezzogiorno, 84132 Salerno, Italy
| | - Stefano Petrini
- National Reference Centre for Infectious Bovine Rhinotracheitis (IBR), Istituto Zooprofilattico Sperimentale Umbria-Marche, “Togo Rosati”, 06126 Perugia, Italy
| | - Giovanna De Matteis
- Research Centre for Animal Production and Aquaculture, Consiglio per la Ricerca in Agricoltura e l’Analisi dell’Economia Agraria (CREA), 00015 Monterotondo, Italy
| | - Alessandra Martucciello
- National Reference Centre for Hygiene and Technologies of Water Buffalo Farming and Productions, Istituto Zooprofilattico Sperimentale del Mezzogiorno, 84132 Salerno, Italy
| |
Collapse
|
10
|
Corleis B, Bastian M, Hoffmann D, Beer M, Dorhoi A. Animal models for COVID-19 and tuberculosis. Front Immunol 2023; 14:1223260. [PMID: 37638020 PMCID: PMC10451089 DOI: 10.3389/fimmu.2023.1223260] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 07/21/2023] [Indexed: 08/29/2023] Open
Abstract
Respiratory infections cause tremendous morbidity and mortality worldwide. Amongst these diseases, tuberculosis (TB), a bacterial illness caused by Mycobacterium tuberculosis which often affects the lung, and coronavirus disease 2019 (COVID-19) caused by the Severe Acute Respiratory Syndrome Coronavirus type 2 (SARS-CoV-2), stand out as major drivers of epidemics of global concern. Despite their unrelated etiology and distinct pathology, these infections affect the same vital organ and share immunopathogenesis traits and an imperative demand to model the diseases at their various progression stages and localizations. Due to the clinical spectrum and heterogeneity of both diseases experimental infections were pursued in a variety of animal models. We summarize mammalian models employed in TB and COVID-19 experimental investigations, highlighting the diversity of rodent models and species peculiarities for each infection. We discuss the utility of non-human primates for translational research and emphasize on the benefits of non-conventional experimental models such as livestock. We epitomize advances facilitated by animal models with regard to understanding disease pathophysiology and immune responses. Finally, we highlight research areas necessitating optimized models and advocate that research of pulmonary infectious diseases could benefit from cross-fertilization between studies of apparently unrelated diseases, such as TB and COVID-19.
Collapse
Affiliation(s)
- Björn Corleis
- Institute of Immunology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald-Insel Riems, Germany
| | - Max Bastian
- Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald-Insel Riems, Germany
| | - Donata Hoffmann
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald-Insel Riems, Germany
| | - Martin Beer
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald-Insel Riems, Germany
| | - Anca Dorhoi
- Institute of Immunology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald-Insel Riems, Germany
- Faculty of Mathematics and Natural Sciences, University of Greifswald, Greifswald, Germany
| |
Collapse
|
11
|
Grandoni F, Fraboni D, Canonico B, Papa S, Buccisano F, Schuberth HJ, Hussen J. Flow Cytometric Identification and Enumeration of Monocyte Subsets in Bovine and Water Buffalo Peripheral Blood. Curr Protoc 2023; 3:e676. [PMID: 36799694 DOI: 10.1002/cpz1.676] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
Monocytes are innate immune system key players with pivotal roles during infection and inflammation. They migrate into tissues and differentiate into myeloid effect cells (macrophages, dendritic cells) which orchestrate inflammatory processes and are interfaces between the innate and adaptive immune responses. Their clinical relevance to health and disease of cattle (Bos taurus) and water buffalo (Bubalus bubalis), two of the most important livestock species, has been highlighted in physiologic (pregnancy) and pathologic (mastitis, metritis, and viral infections) conditions. The existence of three different monocyte subsets in cattle was established by flow cytometry (FC), as follows: classical (cM; CD14++ CD16-/low ), intermediate (intM; CD14++/+ CD16+ ), and non-classical (ncM; CD14-/low CD16++ ) monocytes. FC applications for studying the immune system of cattle and water buffalo still have significant limitations. In this article, we describe some practical approaches to overcome these limitations and, in particular, allow the identification and enumeration of cM, intM, and ncM subpopulations in cattle and buffalo peripheral blood. Indeed, we propose the new procedure lyse/wash/no-centrifugation (L/W/NC) that can be combined with the FC absolute counting procedures and can overcome specific issues of the lyse/no-wash protocols (L/NW). Finally, for the first time, we demonstrated the existence of cM, intM, and ncM monocyte subsets also in the water buffalo, showing some interesting differences with cattle, such as the bubaline cM are mainly CD14+/++ /CD16+ . These subtle differences may influence inflammatory disease regulation in, for example, mastitis and metritis. The upregulation of CD16 expression on cM may reveal different monocyte priming, leading to different functional features of macrophages/dendritic cells in tissues after infection. © 2023 Wiley Periodicals LLC. Basic Protocol: Absolute count of cM, intM, and ncM without compensation Alternate Protocol: Absolute count of cM, intM, and ncM for single laser platform Support Protocol 1: In-house monoclonal antibody labeling using a Pacific Blue™ kit Support Protocol 2: In-house monoclonal antibody labeling using an Alexa Fluor® 647 kit Support Protocol 3: Titration of fluorochrome-conjugated antibodies.
Collapse
Affiliation(s)
- Francesco Grandoni
- CREA-Consiglio per la Ricerca in Agricoltura e l'Analisi dell'Economia Agraria, Centro di ricerca Zootecnia e Acquacoltura (Research Center for Animal Production and Aquaculture), Monterotondo, Italy
| | | | - Barbara Canonico
- Department of Biomolecular Sciences, University of Urbino "Carlo Bo", Urbino, Italy
| | - Stefano Papa
- Department of Biomolecular Sciences, University of Urbino "Carlo Bo", Urbino, Italy
| | - Francesco Buccisano
- Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - Hans-Joachim Schuberth
- Institute for Immunology, University of Veterinary Medicine Foundation, Hanover, Germany
| | - Jamal Hussen
- Department of Microbiology, College of Veterinary Medicine, King Faisal University, Al-Ahsa, Saudi Arabia
| |
Collapse
|
12
|
Barut GT, Kreuzer M, Bruggmann R, Summerfield A, Talker SC. Single-cell transcriptomics reveals striking heterogeneity and functional organization of dendritic and monocytic cells in the bovine mesenteric lymph node. Front Immunol 2023; 13:1099357. [PMID: 36685557 PMCID: PMC9853064 DOI: 10.3389/fimmu.2022.1099357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 12/13/2022] [Indexed: 01/09/2023] Open
Abstract
Dendritic and monocytic cells co-operate to initiate and shape adaptive immune responses in secondary lymphoid tissue. The complexity of this system is poorly understood, also because of the high phenotypic and functional plasticity of monocytic cells. We have sequenced mononuclear phagocytes in mesenteric lymph nodes (LN) of three adult cows at the single-cell level, revealing ten dendritic-cell (DC) clusters and seven monocyte/macrophage clusters with clearly distinct transcriptomic profiles. Among DC, we defined LN-resident subsets and their progenitors, as well as subsets of highly activated migratory DC differing in transcript levels for T-cell attracting chemokines. Our analyses also revealed a potential differentiation path for cDC2, resulting in a cluster of inflammatory cDC2 with close transcriptional similarity to putative DC3 and monocyte-derived DC. Monocytes and macrophages displayed sub-clustering mainly driven by pro- or anti-inflammatory expression signatures, including a small cluster of cycling, presumably self-renewing, macrophages. With this transcriptomic snapshot of LN-derived mononuclear phagocytes, we reveal functional properties and differentiation trajectories in a "command center of immunity", and identify elements that are conserved across species.
Collapse
Affiliation(s)
- Güliz Tuba Barut
- Institute of Virology and Immunology, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Marco Kreuzer
- Interfaculty Bioinformatics Unit and Swiss Institute of Bioinformatics, University of Bern, Bern, Switzerland
| | - Rémy Bruggmann
- Interfaculty Bioinformatics Unit and Swiss Institute of Bioinformatics, University of Bern, Bern, Switzerland
| | - Artur Summerfield
- Institute of Virology and Immunology, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Stephanie C. Talker
- Institute of Virology and Immunology, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| |
Collapse
|
13
|
Wang L, Mei X, Liu X, Guo L, Yang B, Chen R. The Interleukin-33/ST2 Axis Enhances Lung-Resident CD14+ Monocyte Function in Patients with Non-Small Cell Lung Cancer. Immunol Invest 2023; 52:67-82. [PMID: 36218388 DOI: 10.1080/08820139.2022.2130075] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Interleukin-33 (IL-33) binds to its cognate receptor suppression of tumorigenicity 2 (ST2), leading to critical modulatory roles in immune responses during inflammation and cancers. The aim of this study was to investigate the role of IL-33/ST2 signaling in monocyte function in non-small cell lung cancer (NSCLC). Sixty-two NSCLC patients and nineteen controls were enrolled. IL-33 levels and ST2 expression were measured in peripheral blood and bronchoalveolar lavage fluid (BALF) by ELISA and flow cytometry. HLA-DR expression by CD14+ monocytes, granzyme B and proinflammatory cytokine secretion were also investigated in lipopolysaccharide-stimulated cells. CD14+ monocytes purified from BALF in the tumor site were stimulated with IL-33 in vitro, and co-cultured with a lung cancer cell line A549 cells. The cytotoxicity of monocytes with IL-33 stimulation was then assessed. IL-33 levels were lower in the peripheral blood and tumor microenvironment of NSCLC patients. There was no significant difference in peripheral ST2 expression between NSCLC patients and controls. Soluble ST2 levels were increased but membrane-bound ST2 expression in CD14+ monocytes was decreased in tumor microenvironment of NSCLC patients. There were no remarkable differences in either HLA-DR expression or proinflammatory cytokine secretion by circulating CD14+ monocytes between NSCLC patients and controls. CD14+ monocytes in the tumor microenvironment revealed a dysfunctional phenotype, which presented as lower HLA-DR expression and reduced granzyme B and proinflammatory cytokines. A higher concentration of IL-33 stimulation promoted tumor-resident CD14+ monocyte-induced target cell death. The present study indicates that IL-33/ST2 signaling pathway might enhance the activity of tumor-resident CD14+ monocytes in NSCLC.
Collapse
Affiliation(s)
- Lv Wang
- Department of Thoracic Surgery, Xi'an Daxing Hospital, Xi'an, Shaanxi, China
| | - Xingke Mei
- Department of Thoracic Surgery, Xi'an Daxing Hospital, Xi'an, Shaanxi, China
| | - Xiaogang Liu
- Department of Thoracic Surgery, Xi'an Daxing Hospital, Xi'an, Shaanxi, China
| | - Lu Guo
- Department of Thoracic Surgery, Xi'an Daxing Hospital, Xi'an, Shaanxi, China
| | - Bo Yang
- Department of Thoracic Surgery, Xi'an Daxing Hospital, Xi'an, Shaanxi, China
| | - Ren'an Chen
- The Second Department of Internal Medicine, Shaanxi Cancer Hospital, Xi'an, Shaanxi, China
| |
Collapse
|