Shpigel Lab

Recent Publications

Hyperinflammation evokes different antiviral TMPRSS2 and ADAM17 expression responses in human gut xenograft versus host mouse gut although overall genomic responses are similar

Lorna Morris, Einat Nisim-Eliraz, , Schouten, Iftach , François Bergey, , Nigel Dyer, , Hiroshi Momiji, , Nir, Eilam , Marsiano, Noga , Raheleh Sheibani Tezerji, , Yagel, Simcha , Rosenstiel, Philip , Rand, David , dos Santos, Vitor A.P. Martins , Shpigel, Nahum Y. , and , . 2021. Hyperinflammation Evokes Different Antiviral Tmprss2 And Adam17 Expression Responses In Human Gut Xenograft Versus Host Mouse Gut Although Overall Genomic Responses Are Similar. Biorxiv, Pp. 2021.05.09.443289. http://biorxiv.org/content/early/2021/05/10/2021.05.09.443289.abstract. Publisher's Version Abstract
The global spread of the newly emerged severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has led to the pandemic outbreak of coronavirus disease 2019 (COVID-19), an inflammatory disease that is primarily affecting the respiratory system. However, gastrointestinal symptoms in COVID-19 patients suggests that the gut may present another viral target organ. Disease development and severity is dependent on viral interaction with two cell surface human proteins, ACE2 and TMPRSS2, and on antiviral response which may lead to systemic hyperinflammatory syndrome and multiorgan dysfunction. Understanding the host response to SARS-CoV-2 infection and the pathology of the disease will be greatly enhanced by the development of appropriate animal models. Laboratory mice have been the mainstay of therapeutic and vaccine development, however, the virus does not grow in wild type mice and only induced mild disease in transgenic animals expressing human ACE2. As there are known differences between immune response in laboratory mice and humans we evaluated the response of human gut developed as xenografts and host mouse gut following systemic LPS injections as a hyperinflammation model system. The orthologous gene expression levels in the mouse and human gut were highly correlated (Spearman’s rank correlation coefficient: 0.28–0.76) and gene set enrichment analysis of significantly upregulated human and mouse genes revealed that a number of inflammatory and immune response pathways are commonly regulated in the two species. However, species differences were also observed, most importantly, in the inflamed human gut but not in the mouse gut, there was clear upregulation of mRNAs coding for TMPRSS2, ADAM17 and for RIG-I-like receptors, which are involved in the recognition of viruses and in antiviral innate immune response. Moreover, using species-specific immunofluorescence microscopy, we demonstrated the expression and localization of human ACE2 and TMPRSS2 proteins, which are essential elements of the molecular machinery that enables SARS-CoV-2 to infect and replicate in human gut cells. Our findings demonstrate that the intestinal immune response to inflammation in humans and mice are generally very similar. However, certain human-specific diseases, such as COVID-19, can only be successfully studied in an experimental model of human tissue, such as the gut xenograft.Competing Interest StatementThe authors have declared no competing interest.

Homeostasis of mucosal glial cells in human gut is independent of microbiota

Timna Inlender, Nissim-Eliraz, Einat , Stavely, Rhian , Hotta, Ryo , Goldstein, Allan M, Yagel, Simcha , Gutnick, Michael J, and Shpigel, Nahum Y. 2021. Homeostasis Of Mucosal Glial Cells In Human Gut Is Independent Of Microbiota. Sci Rep, 11, 1, Pp. 12796. doi:10.1038/s41598-021-92384-9. Abstract
In mammals, neural crest cells populate the gut and form the enteric nervous system (ENS) early in embryogenesis. Although the basic ENS structure is highly conserved across species, we show important differences between mice and humans relating to the prenatal and postnatal development of mucosal enteric glial cells (mEGC), which are essential ENS components. We confirm previous work showing that in the mouse mEGCs are absent at birth, and that their appearance and homeostasis depends on postnatal colonization by microbiota. In humans, by contrast, a network of glial cells is already present in the fetal gut. Moreover, in xenografts of human fetal gut maintained for months in immuno-compromised mice, mEGCs persist following treatment with antibiotics that lead to the disappearance of mEGCs from the gut of the murine host. Single cell RNAseq indicates that human and mouse mEGCs differ not only in their developmental dynamics, but also in their patterns of gene expression.

NF-kappa-B activation unveils the presence of inflammatory hotspots in human gut xenografts

Einat Nissim-Eliraz, Nir, Eilam , Marsiano, Noga , Yagel, Simcha , and Shpigel, Nahum Y. 2021. Nf-Kappa-B Activation Unveils The Presence Of Inflammatory Hotspots In Human Gut Xenografts. Plos One, 16, 5, Pp. e0243010. doi:10.1371/journal.pone.0243010. Abstract
The single-epithelial cell layer of the gut mucosa serves as an essential barrier between the host and luminal microflora and plays a major role in innate immunity against invading pathogens. Nuclear factor kB (NF-κB), a central component of the cellular signaling machinery, regulates immune response and inflammation. NF-κB proteins are activated by signaling pathways downstream to microbial recognition receptors and cytokines receptors. Highly regulated NF-κB activity in intestinal epithelial cells (IEC) is essential for normal gut homeostasis; dysregulated activity has been linked to a number of disease states, including inflammatory bowel diseases (IBD) such as Crohn's Disease (CD). Our aim was to visualize and quantify spatial and temporal dynamics of NF-κB activity in steady state and inflamed human gut. Lentivirus technology was used to transduce the IEC of human gut xenografts in SCID mice with a NF-κB luminescence reporter system. NF-κB signaling was visualized and quantified using low resolution, intravital imaging of the whole body and high resolution, immunofluorescence microscopic imaging of the tissues. We show that NF-κB is activated in select subset of IEC with low "leaky" NF-κB activity. These unique inflammatory epithelial cells are clustered in the gut into discrete hotspots of NF-κB activity that are visible in steady state and selectively activated by systemic LPS and human TNFα or luminal bacteria. The presence of inflammatory hotspots in the normal and inflamed gut might explain the patchy mucosal lesions characterizing CD and thus could have important implications for diagnosis and therapy.

The role of O-polysaccharide chain and complement resistance of Escherichia coli in mammary virulence

Hagit Salamon, Nissim-Eliraz, Einat , Ardronai, Oded , Nissan, Israel , and Shpigel, Nahum Y. . 2020. The Role Of O-Polysaccharide Chain And Complement Resistance Of Escherichia Coli In Mammary Virulence. Vet Res, 51, 1, Pp. 77. https://doi.org/10.1186/s13567-020-00804-x. Publisher's Version Abstract
Mastitis, inflammation of the mammary gland, is a common disease of dairy animals. The disease is caused by bacterial infection ascending through the teat canal and mammary pathogenic Escherichia coli (MPEC) are common etiology. In the first phase of infection, virulence mechanisms, designated as niche factors, enable MPEC bacteria to resist innate antimicrobial mechanisms, replicate in milk, and to colonize the mammary gland. Next, massive replication of colonizing bacteria culminates in a large biomass of microbe-associated molecular patterns (MAMPs) recognized by pattern recognition receptors (PRRs) such as toll-like receptors (TLRs) mediating inflammatory signaling in mammary alveolar epithelial cells (MAEs) and macrophages. Bacterial lipopolysaccharides (LPSs), the prototypical class of MAMPs are sufficient to elicit mammary inflammation mediated by TLR4 signaling and activation of nuclear factor kB (NF-kB), the master regulator of inflammation. Using in vivo mastitis model, in low and high complements mice, and in vitro NF-kB luminescence reporter system in MAEs, we have found that the smooth configuration of LPS O-polysaccharides in MPEC enables the colonizing organisms to evade the host immune response by reducing inflammatory response and conferring resistance to complement. Screening a collection of MPEC field strains, we also found that all strains were complement resistant and 94% (45/48) were smooth. These results indicate that the structure of LPS O-polysaccharides chain is important for the pathogenesis of MPEC mastitis and provides protection against complement-mediated killing. Furthermore, we demonstrate a role for complement, a key component of innate immunity, in host-microbe interactions of the mammary gland.

NF-kappa-B activation unveils the presence of inflammatory hotspots in human gut xenografts

Einat Nissim-Eliraz, Nir, Eilam , Marsiano, Noga , Yagel, Simcha , and Shpigel, Nahum Y. 2020. Nf-Kappa-B Activation Unveils The Presence Of Inflammatory Hotspots In Human Gut Xenografts. Biorxiv, Pp. 2020.07.23.212621. doi:10.1101/2020.07.23.212621. Publisher's Version Abstract
The single-epithelial cell layer of the gut mucosa serves as an essential barrier between the host and luminal microflora and plays a major role in innate immunity against invading pathogens. Nuclear factor kB (NF-kB), a central component of the cellular signaling machinery, regulates immune response and inflammation. NF-kB proteins are activated by signaling pathways downstream to microbial recognition receptors and cytokines receptors. Highly regulated NF-kB activity in intestinal epithelial cells (IEC) is essential for normal gut homeostasis; dysregulated activity has been linked to a number of disease states, including inflammatory bowel diseases (IBD) such as Crohns Disease (CD). Our aim was to visualize and quantify spatial and temporal dynamics of NF-kB activity in steady state and inflamed human gut. Lentivirus technology was used to transduce the IEC of human gut xenografts in SCID mice with a NF-kB luminescence reporter system. NF-kB signaling was visualized and quantified using low resolution, intravital imaging of the whole body and high resolution, immunofluorescence microscopic imaging of the tissues. We show that NF-kB is activated in select subset of IEC with low leaky NF-kB activity. These unique inflammatory epithelial cells are clustered in the gut into discrete hotspots of NF-kB activity that are visible in steady state and selectively activated by systemic LPS and human TNFa; or luminal bacteria. The presence of inflammatory hotspots in the normal and inflamed gut might explain the patchy mucosal lesions characterizing CD and thus could have important implications for diagnosis and therapy.Competing Interest StatementThe authors have declared no competing interest.
More