1
|
Yeoh WJ, Krebs P. SHIP1 and its role for innate immune regulation-Novel targets for immunotherapy. Eur J Immunol 2023; 53:e2350446. [PMID: 37742135 DOI: 10.1002/eji.202350446] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 08/03/2023] [Accepted: 09/21/2023] [Indexed: 09/25/2023]
Abstract
Phosphoinositide-3-kinase/AKT (PI3K/AKT) signaling plays key roles in the regulation of cellular activity in both health and disease. In immune cells, this PI3K/AKT pathway is critically regulated by the phosphoinositide phosphatase SHIP1, which has been reported to modulate the function of most immune subsets. In this review, we summarize our current knowledge of SHIP1 with a focus on innate immune cells, where we reflect on the most pertinent aspects described in the current literature. We also present several small-molecule agonists and antagonists of SHIP1 developed over the last two decades, which have led to improved outcomes in several preclinical models of disease. We outline these promising findings and put them in relation to human diseases with unmet medical needs, where we discuss the most attractive targets for immune therapies based on SHIP1 modulation.
Collapse
Affiliation(s)
- Wen Jie Yeoh
- Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Philippe Krebs
- Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
| |
Collapse
|
2
|
Kesserwan S, Sadagurski M, Mao L, Klueh U. Mast Cell Deficiency in Mice Attenuates Insulin Phenolic Preservative-Induced Inflammation. Biomedicines 2023; 11:2258. [PMID: 37626754 PMCID: PMC10452641 DOI: 10.3390/biomedicines11082258] [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: 07/18/2023] [Revised: 08/04/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
One major obstacle that limits the lifespan of insulin infusion pumps is surmounting the tissue site reaction at the device implantation site. All commercial insulin formulations contain insulin phenolic preservatives (IPPs) designed to ensure insulin protein stability and prolong shelf-life. However, our laboratory demonstrated that these preservatives are cytotoxic and induce inflammation. Mature mast cells (MCs) reside in cutaneous tissue and are one of the first responders to an epidermal breach. Upon activation, MCs release proinflammatory and immunomodulatory prepacked mediators that exacerbate these inflammatory reactions. Thus, we hypothesized that once the epidermis is breached, cutaneous MCs are triggered inciting the inflammatory response to IPP-induced inflammation. This hypothesis was pursued utilizing our modified in vivo mouse air pouch model, including a c-kit dependent (C57BL/6J-kitW-sh/W-sh) and a c-kit independent (Cpa3-Cre; Mcl-1fl/fl) MC-deficient mouse model. Leukocytes were quantified in the mouse air pouch lavage fluid following flow cytometry analysis for IPP infusion under three different states, insulin-containing phenolic preservatives (Humalog®), insulin preservatives alone, and normal saline as a control. The air pouch wall was assessed using histopathological evaluations. Flow cytometry analysis demonstrated a statistically significant difference in inflammatory cell recruitment for both MC-deficient mouse models when compared to the control strain including infused control saline. Significantly less inflammation was observed at the site of infusion for the MC-deficient strains compared to the control strain. Overall, concordant results were obtained in both mouse types, C57Bl6-kitW-sh/W-sh and Cpa3-Cre; Mcl-1fl/fl. These findings in multiple model systems support the conclusion that MCs have important or possible unique roles in IPP-induced inflammation.
Collapse
Affiliation(s)
| | | | | | - Ulrike Klueh
- Integrative Biosciences Center (IBio), Wayne State University, Detroit, MI 48202, USA; (S.K.); (M.S.)
| |
Collapse
|
3
|
Rose N, Holdermann S, Callegari I, Kim H, Fruh I, Kappos L, Kuhle J, Müller M, Sanderson NSR, Derfuss T. Receptor clustering and pathogenic complement activation in myasthenia gravis depend on synergy between antibodies with multiple subunit specificities. Acta Neuropathol 2022; 144:1005-1025. [PMID: 36074148 PMCID: PMC9547806 DOI: 10.1007/s00401-022-02493-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 08/17/2022] [Accepted: 09/02/2022] [Indexed: 01/26/2023]
Abstract
Myasthenia gravis is an autoimmune disorder defined by muscle weakness and fatigability associated with antibodies against proteins of the neuromuscular junction (NMJ). The most common autoantibody target is the acetylcholine receptor (AChR). Three mechanisms have been postulated by which autoantibodies might interfere with neurotransmission: direct antagonism of the receptor, complement-mediated destruction of the postsynaptic membrane, and enhanced internalization of the receptor. It is very likely that more than one of these mechanisms act in parallel. Dissecting the mechanisms of autoantibody-mediated pathology requires patient-derived, monoclonal antibodies. Using membrane antigen capture activated cell sorting (MACACS), we isolated AChR-specific B cells from patients with myasthenia gravis, and produced six recombinant antibodies. All AChR-specific antibodies were hypermutated, including isotypes IgG1, IgG3, and IgG4, and recognized different subunits of the AChR. Despite clear binding, none of the individual antibodies showed significant antagonism of the AChR measured in an in vitro neuromuscular synapse model, or AChR-dependent complement activation, and they did not induce myasthenic signs in vivo. However, combinations of antibodies induced strong complement activation in vitro, and severe weakness in a passive transfer myasthenia gravis rat model, associated with NMJ destruction and complement activation in muscle. The strongest complement activation was mediated by combinations of antibodies targeting disparate subunits of the AChR, and such combinations also induced the formation of large clusters of AChR on the surface of live cells in vitro. We propose that synergy between antibodies of different epitope specificities is a fundamental feature of this disease, and possibly a general feature of complement-mediated autoimmune diseases. The importance of synergistic interaction between antibodies targeting different subunits of the receptor can explain the well-known discrepancy between serum anti-AChR titers and clinical severity, and has implications for therapeutic strategies currently under investigation.
Collapse
Affiliation(s)
- Natalie Rose
- Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
- Neurologic Clinic and Policlinic and MS Center, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Sebastian Holdermann
- Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
- Neurologic Clinic and Policlinic and MS Center, University Hospital Basel, University of Basel, Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience (RC2NB), University Hospital and University of Basel, Basel, Switzerland
| | - Ilaria Callegari
- Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
- Neurologic Clinic and Policlinic and MS Center, University Hospital Basel, University of Basel, Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience (RC2NB), University Hospital and University of Basel, Basel, Switzerland
| | - Hyein Kim
- Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
- Neurologic Clinic and Policlinic and MS Center, University Hospital Basel, University of Basel, Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience (RC2NB), University Hospital and University of Basel, Basel, Switzerland
| | - Isabelle Fruh
- Chemical Biology and Therapeutics, Novartis Institutes for BioMedical Research, 4002, Basel, Switzerland
| | - Ludwig Kappos
- Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
- Neurologic Clinic and Policlinic and MS Center, University Hospital Basel, University of Basel, Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience (RC2NB), University Hospital and University of Basel, Basel, Switzerland
| | - Jens Kuhle
- Neurologic Clinic and Policlinic and MS Center, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Matthias Müller
- Chemical Biology and Therapeutics, Novartis Institutes for BioMedical Research, 4002, Basel, Switzerland
| | - Nicholas S R Sanderson
- Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland.
- Neurologic Clinic and Policlinic and MS Center, University Hospital Basel, University of Basel, Basel, Switzerland.
- Research Center for Clinical Neuroimmunology and Neuroscience (RC2NB), University Hospital and University of Basel, Basel, Switzerland.
| | - Tobias Derfuss
- Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
- Neurologic Clinic and Policlinic and MS Center, University Hospital Basel, University of Basel, Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience (RC2NB), University Hospital and University of Basel, Basel, Switzerland
| |
Collapse
|
4
|
Understanding human mast cells: lesson from therapies for allergic and non-allergic diseases. Nat Rev Immunol 2022; 22:294-308. [PMID: 34611316 DOI: 10.1038/s41577-021-00622-y] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/19/2021] [Indexed: 02/07/2023]
Abstract
Mast cells have crucial roles in allergic and other inflammatory diseases. Preclinical approaches provide circumstantial evidence for mast cell involvement in many diseases, but these studies have major limitations - for example, there is still a lack of suitable mouse models for some mast cell-driven diseases such as urticaria. Some approaches for studying mast cells are invasive or can induce severe reactions, and very few mediators or receptors are specific for mast cells. Recently, several drugs that target human mast cells have been developed. These include monoclonal antibodies and small molecules that can specifically inhibit mast cell degranulation via key receptors (such as FcεRI), that block specific signal transduction pathways involved in mast cell activation (for example, BTK), that silence mast cells via inhibitory receptors (such as Siglec-8) or that reduce mast cell numbers and prevent their differentiation by acting on the mast/stem cell growth factor receptor KIT. In this Review, we discuss the existing and emerging therapies that target mast cells, and we consider how these treatments can help us to understand mast cell functions in disease.
Collapse
|
5
|
Fu W, Chen C, Xie Q, Gu S, Tao S, Xue W. Pediococcus acidilactici Strain Alleviates Gluten-Induced Food Allergy and Regulates Gut Microbiota in Mice. Front Cell Infect Microbiol 2022; 12:845142. [PMID: 35531345 PMCID: PMC9072736 DOI: 10.3389/fcimb.2022.845142] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 03/23/2022] [Indexed: 11/13/2022] Open
Abstract
Wheat flour, the most important source of food globally, is also one of the most common causative agents of food allergy. Wheat gluten protein, which accounts for 80% of the total wheat protein, is a major determinant of important wheat-related disorders. In this study, the effects of Pediococcus acidilactici XZ31 against gluten-induced allergy were investigated in a mouse model. The oral administration of P. acidilactici XZ31 attenuated clinical and intestinal allergic responses in allergic mice. Further results showed that P. acidilactici XZ31 regulated Th1/Th2 immune balance toward Th1 polarization, which subsequently induced a reduction in gluten-specific IgE production. We also found that P. acidilactici XZ31 modulated gut microbiota homeostasis by balancing the Firmicutes/Bacteroidetes ratio and increasing bacterial diversity and the abundance of butyrate-producing bacteria. Specifically, the abundance of Firmicutes and Erysipelotrichaceae is positively correlated with concentrations of gluten-specific IgE and may act as a fecal biomarker for diagnosis. The evidence for the role of P. acidilactici XZ31 in alleviating gluten-induced allergic responses sheds light on the application of P. acidilactici XZ31 in treating wheat allergy.
Collapse
Affiliation(s)
- Wenhui Fu
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Chen Chen
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Qiang Xie
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Shimin Gu
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Sha Tao
- College of Information and Electrical Engineering, China Agricultural University, Beijing, China
| | - Wentong Xue
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
- *Correspondence: Wentong Xue, ;
| |
Collapse
|
6
|
Himelreich-Perić M, Katušić-Bojanac A, Hohšteter M, Sinčić N, Mužić-Radović V, Ježek D. Mast Cells in the Mammalian Testis and Epididymis-Animal Models and Detection Methods. Int J Mol Sci 2022; 23:ijms23052547. [PMID: 35269690 PMCID: PMC8909951 DOI: 10.3390/ijms23052547] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 02/21/2022] [Accepted: 02/23/2022] [Indexed: 02/04/2023] Open
Abstract
Mast cells (MCs) are an evolutionary well-conserved type of cells, mediating and modulating allergic responses in innate immunity and tissue remodeling after chronic inflammation. Among other tissues, they inhabit both the testis and epididymis. In the testis, MCs usually appear in the interstitial compartment in humans, but not in other standard experimental models, like rats and mice. MCs seem to be responsible for testicular tissue fibrosis in different causes of infertility. Although experimental animal models follow the effect on MC activation or penetration to the interstitial tissue like in humans to some extent, there is an inconsistency in the available literature regarding experimental design, animal strain, and detection methods used. This comprehensive review offers an insight into the literature on MCs in mammalian testes and epididymides. We aimed to find the most suitable model for research on MC and offer recommendations for future experimental designs. When using in vivo animal models, tunica albuginea incorporation and standard histological assessment need to be included. Domesticated boar strains kept in modified controlled conditions exhibit the highest similarity to the MC distribution in the human testis. 3D testicular models are promising but need further fine-tuning to become a valid model for MC investigation.
Collapse
Affiliation(s)
- Marta Himelreich-Perić
- Scientific Centre of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (A.K.-B.); (N.S.); (D.J.)
- Department of Biology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
- Correspondence:
| | - Ana Katušić-Bojanac
- Scientific Centre of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (A.K.-B.); (N.S.); (D.J.)
- Department of Biology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Marko Hohšteter
- Department of Veterinary Pathology, Faculty of Veterinary Medicine, University of Zagreb, 10000 Zagreb, Croatia;
| | - Nino Sinčić
- Scientific Centre of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (A.K.-B.); (N.S.); (D.J.)
- Department of Biology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Vedrana Mužić-Radović
- Hospital for Medical Rehabilitation of the Health and Lung Diseases and Rheumatism “Thalassotherapia-Opatija”, 51410 Opatija, Croatia;
| | - Davor Ježek
- Scientific Centre of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (A.K.-B.); (N.S.); (D.J.)
- Department of Histology and Embryology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| |
Collapse
|
7
|
Dahlin JS, Maurer M, Metcalfe DD, Pejler G, Sagi‐Eisenberg R, Nilsson G. The ingenious mast cell: Contemporary insights into mast cell behavior and function. Allergy 2022; 77:83-99. [PMID: 33955017 DOI: 10.1111/all.14881] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 04/15/2021] [Accepted: 04/16/2021] [Indexed: 12/14/2022]
Abstract
Mast cells are (in)famous for their role in allergic diseases, but the physiological and pathophysiological roles of this ingenious cell are still not fully understood. Mast cells are important for homeostasis and surveillance of the human system, recognizing both endogenous and exogenous agents, which induce release of a variety of mediators acting on both immune and non-immune cells, including nerve cells, fibroblasts, endothelial cells, smooth muscle cells, and epithelial cells. During recent years, clinical and experimental studies on human mast cells, as well as experiments using animal models, have resulted in many discoveries that help decipher the function of mast cells in health and disease. In this review, we focus particularly on new insights into mast cell biology, with a focus on mast cell development, recruitment, heterogeneity, and reactivity. We also highlight the development in our understanding of mast cell-driven diseases and discuss the development of novel strategies to treat such conditions.
Collapse
Affiliation(s)
- Joakim S. Dahlin
- Division of Immunology and Allergy Department of Medicine Karolinska Institutet Karolinska University Hospital Stockholm Sweden
| | - Marcus Maurer
- Department of Dermatology and Allergy Dermatological Allergology Allergie‐Centrum‐Charité Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt‐Universität zu Berlin, Berlin Institute of Health Berlin Germany
| | - Dean D. Metcalfe
- Mast Cell Biology Section Laboratory of Allergic Diseases NIAID, NIH Bethesda MD USA
| | - Gunnar Pejler
- Department of Medical Biochemistry and Microbiology Uppsala University Uppsala Sweden
- Department of Anatomy, Physiology and Biochemistry Swedish University of Agricultural Sciences Uppsala Sweden
| | - Ronit Sagi‐Eisenberg
- Department of Cell and Developmental Biology Sackler Faculty of Medicine Tel Aviv University Tel Aviv Israel
| | - Gunnar Nilsson
- Division of Immunology and Allergy Department of Medicine Karolinska Institutet Karolinska University Hospital Stockholm Sweden
- Department of Medical Sciences Uppsala University Uppsala Sweden
| |
Collapse
|
8
|
Chin HS, Fu NY. Physiological Functions of Mcl-1: Insights From Genetic Mouse Models. Front Cell Dev Biol 2021; 9:704547. [PMID: 34336857 PMCID: PMC8322662 DOI: 10.3389/fcell.2021.704547] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 06/14/2021] [Indexed: 01/27/2023] Open
Abstract
The ability to regulate the survival and death of a cell is paramount throughout the lifespan of a multicellular organism. Apoptosis, a main physiological form of programmed cell death, is regulated by the Bcl-2 family proteins that are either pro-apoptotic or pro-survival. The in vivo functions of distinct Bcl-2 family members are largely unmasked by genetically engineered murine models. Mcl-1 is one of the two Bcl-2 like pro-survival genes whose germline deletion causes embryonic lethality in mice. Its requisite for the survival of a broad range of cell types has been further unraveled by using conditional and inducible deletion murine model systems in different tissues or cell lineages and at distinct developmental stages. Moreover, genetic mouse cancer models have also demonstrated that Mcl-1 is essential for the survival of multiple tumor types. The MCL-1 locus is commonly amplified across various cancer types in humans. Small molecule inhibitors with high affinity and specificity to human MCL-1 have been developed and explored for the treatment of certain cancers. To facilitate the pre-clinical studies of MCL-1 in cancer and other diseases, transgenic mouse models over-expressing human MCL-1 as well as humanized MCL-1 mouse models have been recently engineered. This review discusses the current advances in understanding the physiological roles of Mcl-1 based on studies using genetic murine models and its critical implications in pathology and treatment of human diseases.
Collapse
Affiliation(s)
- Hui San Chin
- Programme in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - Nai Yang Fu
- Programme in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore.,Department of Physiology, National University of Singapore, Singapore, Singapore
| |
Collapse
|
9
|
Sasaki H, Imanishi M, Fujikura D, Sugiyama M, Tanimoto K, Mochiji Y, Takahashi Y, Hiura K, Watanabe M, Kashimoto T, Nakano K, Okamura T, Sasaki N. New inducible mast cell-deficient mouse model (Mcpt5/Cma1 DTR). Biochem Biophys Res Commun 2021; 551:127-132. [PMID: 33725574 DOI: 10.1016/j.bbrc.2021.03.025] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 03/05/2021] [Indexed: 02/03/2023]
Abstract
Mast cell-deficient mice are helpful for understanding the roles of mast cells in vivo. To date, a dozen mouse models for mast cell deficiency have been reported. However, mice with a specific depletion of all populations of mast cells have not been reported. We generated knock-in mice, termed Mcpt5/Cma1DTR mice, expressing human diphtheria toxin A (DT) receptor under the endogenous promoter of Mcpt5 (also known as Cma1), which encodes mouse mast cell protease-5. Flow cytometry and histological analysis showed that intraperitoneal injection of DT induced almost complete depletion of mast cells in heterozygote Mcpt5/Cma1DTR/+ mice. The deletion rates of mast cells in peritoneal cavity, mesentery, abdominal skin, ear skin, and glandular stomach were 99.9%, 100%, 98.7%, 97.7%, and 100%, respectively. Passive cutaneous anaphylaxis reaction also revealed mast cell deficiency in ear skin after DT treatment. Other than mast cells, a small percentage of marginal zone B cells in Mcpt5/Cma1DTR/+ mice were killed by DT treatment. In conclusion, the Mcpt5/Cma1DTR/+ mouse model is valuable for achieving conditional depletion of all populations of mast cells without inducing a marked reduction in other cells.
Collapse
Affiliation(s)
- Hayato Sasaki
- School of Veterinary Medicine, Kitasato University, Towada, 034-8628, Japan
| | - Madoka Imanishi
- School of Veterinary Medicine, Kitasato University, Towada, 034-8628, Japan
| | - Daisuke Fujikura
- School of Veterinary Medicine, Kitasato University, Towada, 034-8628, Japan
| | - Makoto Sugiyama
- School of Veterinary Medicine, Kitasato University, Towada, 034-8628, Japan
| | - Kyosuke Tanimoto
- School of Veterinary Medicine, Kitasato University, Towada, 034-8628, Japan
| | - Yohei Mochiji
- School of Veterinary Medicine, Kitasato University, Towada, 034-8628, Japan
| | - Yuki Takahashi
- School of Veterinary Medicine, Kitasato University, Towada, 034-8628, Japan
| | - Koki Hiura
- School of Veterinary Medicine, Kitasato University, Towada, 034-8628, Japan
| | - Masaki Watanabe
- School of Veterinary Medicine, Kitasato University, Towada, 034-8628, Japan
| | | | - Kenta Nakano
- Department of Laboratory Animal Medicine, Research Institute, National Center for Global Health and Medicine, Tokyo, 162-8655, Japan
| | - Tadashi Okamura
- Department of Laboratory Animal Medicine, Research Institute, National Center for Global Health and Medicine, Tokyo, 162-8655, Japan
| | - Nobuya Sasaki
- School of Veterinary Medicine, Kitasato University, Towada, 034-8628, Japan.
| |
Collapse
|