Try a new search

Format these results:

Searched for:

person:korals01

in-biosketch:yes

Total Results:

118


Malignant T cells induce skin barrier defects through cytokine-mediated JAK/STAT signalling in cutaneous T-cell lymphoma

Gluud, Maria; Pallesen, Emil Marek Heymans; Buus, Terkild B; Gjerdrum, Lise M R; Lindahl, Lise M; Kamstrup, Maria R; Bzorek, Michael; Danielsen, Maria; Bech, Rikke; Monteiro, Madalena N; Blümel, Edda; Willerslev-Olsen, Andreas; Lykkebo-Velløe, Anders; Vadivel, Chella Krishna; Krejsgaard, Thorbjørn; Bonefeld, Charlotte M; Geisler, Carsten; Becker, Jürgen C; Koralov, Sergei B; Iversen, Lars; Litman, Thomas; Woetmann, Anders; Odum, Niels
Cutaneous T-cell lymphoma (CTCL) is a devastating lymphoid malignancy characterised by accumulation of malignant T cells in the dermis and epidermis. Skin lesions cause serious symptoms hampering the quality of life and are entry sites for bacterial infection - a major cause of morbidity and mortality in advanced disease. What drives the pathological processes that compromise the skin barrier remains unknown. Here, we report on increased transepidermal water loss and compromised expression of skin barrier proteins filaggrin and filaggrin-2 in areas adjacent to TOX positive T cells in CTCL skin lesions. Malignant T cells secrete mediators (including cytokines such as IL-13, IL-22 and Oncostatin M) that activate STAT3 signalling and downregulate filaggrin and filaggrin-2 expression in human keratinocytes and reconstructed human epithelium. Consequently, repression of filaggrins could be counteracted by a cocktail of antibodies targeting these cytokines/receptors, by siRNA-mediated knockdown of JAK1/STAT3, and by JAK1 inhibitors. Notably, we show that treatment with a clinically approved JAK inhibitor, Tofacitinib, increases filaggrin expression in lesional skin from mycosis fungoides patients. Taken together, these findings indicate that malignant T cells secrete cytokines, which induce skin barrier defects through a JAK1/STAT3 dependent mechanism. As clinical grade JAK inhibitors largely abrogate the negative effect of malignant T cells on skin barrier proteins, our findings suggest that such inhibitors provide novel treatment options for CTCL patients with advanced disease and a compromised skin barrier.
PMID: 36122387
ISSN: 1528-0020
CID: 5335292

Influence of the early-life gut microbiota on the immune responses to an inhaled allergen

Borbet, Timothy C; Pawline, Miranda B; Zhang, Xiaozhou; Wipperman, Matthew F; Reuter, Sebastian; Maher, Timothy; Li, Jackie; Iizumi, Tadasu; Gao, Zhan; Daniele, Megan; Taube, Christian; Koralov, Sergei B; Müller, Anne; Blaser, Martin J
Antibiotics, among the most used medications in children, affect gut microbiome communities and metabolic functions. These changes in microbiota structure can impact host immunity. We hypothesized that early-life microbiome alterations would lead to increased susceptibility to allergy and asthma. To test this, mouse pups between postnatal days 5-9 were orally exposed to water (control) or to therapeutic doses of azithromycin or amoxicillin. Later in life, these mice were sensitized and challenged with a model allergen, house dust mite (HDM), or saline. Mice with early-life azithromycin exposure that were challenged with HDM had increased IgE and IL-13 production by CD4+ T cells compared to unexposed mice; early-life amoxicillin exposure led to fewer abnormalities. To test that the microbiota contained the immunological cues to alter IgE and cytokine production after HDM challenge, germ-free mice were gavaged with fecal samples of the antibiotic-perturbed microbiota. Gavage of adult germ-free mice did not result in altered HDM responses, however, their offspring, which acquired the antibiotic-perturbed microbiota at birth showed elevated IgE levels and CD4+ cytokines in response to HDM, and altered airway reactivity. These studies indicate that early-life microbiota composition can heighten allergen-driven Th2/Th17 immune pathways and airway responses in an age-dependent manner.
PMID: 35842561
ISSN: 1935-3456
CID: 5280022

Oncogenic fusions JAK up CD8+ cytotoxic CTCL [Comment]

Buus, Terkild B; Koralov, Sergei B
PMID: 34882212
ISSN: 1528-0020
CID: 5107802

Robust immune responses are observed after one dose of BNT162b2 mRNA vaccine dose in SARS-CoV-2 experienced individuals

Samanovic, Marie I; Cornelius, Amber R; Gray-Gaillard, Sophie L; Allen, Joseph Richard; Karmacharya, Trishala; Wilson, Jimmy P; Wesley Hyman, Sara; Tuen, Michael; Koralov, Sergei B; Mulligan, Mark J; Sedaghat Herati, Ramin
[Figure: see text].
PMID: 34874183
ISSN: 1946-6242
CID: 5086932

A Transgenic Murine Model Expressing Hyperactive STAT3 Recapitulates the Features of MDS/AML [Meeting Abstract]

Rivera, B L; Gordon, S; Aluri, S; Shi, Y; Chakraborty, S; Choudhary, G S; Shapiro, L C; Pradhan, K; Frank, D A; Koralov, S B; Verma, A; Shastri, A
Myelodysplastic syndromes (MDS) are clonal, myeloid malignancies that emerge and progress due to the expansion of disease-initiating aberrant hematopoietic stem cells that can evolve into Acute Myeloid Leukemia (AML). FDA approved therapies such as the recently approved Bcl-2 inhibitor venetoclax, FLT3 inhibitors, among others, have moved the field forward in newly diagnosed MDS/AML. However, relapsed/refractory (R/R) disease, as well as leukemic transformation post-MDS continues to have a poor prognosis. A pool of hematopoietic stem and progenitor cells (HSPCs) escape chemotherapy, proliferate during disease remission, and causes relapse partly in effect due to signaling effector mutations. It is imperative, for future therapeutic agents, to target these HSPCs populations to achieve a durable remission for aggressive myeloid malignancies. There is an urgent need to develop mouse models that recapitulate human disease for the study of pathogenesis and drug development in these disorders. Signal transducer and activator of transcription 3 (STAT3) belongs to the STAT family of transcription factors that are inappropriately activated in several malignancies. Our preliminary data indicates that STAT3 is overexpressed in MDS and AML stem cells and is associated with an adverse prognosis in a large cohort of patients. (Shastri et al, JCI 2018). We have successfully demonstrated that a selective antisense oligonucleotide inhibitor of STAT3, Danvatirsen, is rapidly incorporated into MDS/AML HSPCs and induces selective apoptosis and downregulation of STAT3 in these cells in comparison with healthy control HSPCs. To determine the role of STAT3 in the initiation of myeloid malignancies, a murine model was generated by crossing R26STAT3C stopfl/fl mice with vavCre transgenic mice. In this model, a hyperactive version of STAT3, STAT3C, is knocked into the Rosa26 locus with an upstream floxed stop cassette (R26STAT3C stopfl). Excision of the stop cassette by Cre recombinase leads to expression of a flag-tagged STAT3C protein and concomitant expression of EGFP in hematopoietic cells. GFP expression allows tracking of cells in which the floxed stop/Neo cassette is deleted and STAT3C is expressed. STAT3C-vavCre double transgenic mice were validated by GFP expression in HSPCs and differentiated hematopoietic cells. The STAT3C-vavCre mice developed ruffled fur, a hunched phenotype and weight-loss by five months of age. CBC analysis of STAT3C-vavCre mice shows a proliferative phenotype reminiscent of high-risk MDS/AML with higher WBC & platelet counts and lower hemoglobin (Figure 1A). Review of the peripheral smear showed an increase in granulocytic precursors that are likely leukemic blasts (Fig 1E). In addition, STAT3C-vavCre mice developed massive splenomegaly (Figure 1B). HSC lineage analysis by FACS showed the presence of GFP positive cells (Figure 1C) with increased expansion of the MPP and HSC compartment compared to controls, suggesting a stem and progenitor phenotype (Figure 1D). Murine myeloid colony assays showed larger colonies in the STAT3C-vavCre mice compared to controls. At this time, single cell RNA sequencing, and bulk RNA sequencing are being performed and will be used to further characterize the phenotype of the STAT3C-vavCre transgenic mice in addition to bone marrow and splenic aspirates & biopsies. Through the generation of a STAT3C-vavCre mouse model, that recapitulates the features of MDS/AML, we aim to further our understanding of the molecular mechanisms and pathways that play an important role in MDS to AML transformation and will help us identify downstream mediators of this event that can be therapeutically targeted. We would also like to use this murine model as an ideal substrate for preclinical studies of STAT3 targeting therapies in hematologic malignancies such as previously reported antisense inhibitors of STAT3 and STAT3 degraders. [Formula presented] Disclosures: Frank: Roche Genentech: Research Funding; Kymera: Consultancy, Research Funding; Revitope: Consultancy; Vigeo: Consultancy. Verma: Throws Exception: Current equity holder in publicly-traded company; BMS: Research Funding; GSK: Research Funding; Acceleron: Consultancy; Incyte: Research Funding; Stelexis: Current equity holder in publicly-traded company; Medpacto: Research Funding; Curis: Research Funding; Eli Lilly: Research Funding; Celgene: Consultancy; Stelexis: Consultancy, Current equity holder in publicly-traded company; Novartis: Consultancy. Shastri: Kymera Therapeutics: Research Funding; GLC: Consultancy; Guidepoint: Consultancy; Onclive: Honoraria.
Copyright
EMBASE:2016075972
ISSN: 0006-4971
CID: 5099072

MicroRNA regulation of B cell receptor signaling

Borbet, Timothy C; Hines, Marcus J; Koralov, Sergei B
B lymphocytes play a central role in host immune defense. B cell receptor (BCR) signaling regulates survival, proliferation, and differentiation of B lymphocytes. Signaling through the BCR signalosome is a multi-component cascade that is tightly regulated and is important in the coordination of B cell differentiation and function. At different stages of development, B cells that have BCRs recognizing self are eliminated to prevent autoimmunity. microRNAs (miRNAs) are small single-stranded non-coding RNAs that contribute to post-transcriptional regulation of gene expression and have been shown to orchestrate cell fate decisions through the regulation of lineage-specific transcriptional profiles. Studies have identified miRNAs to be crucial for B cell development in the bone marrow and their subsequent population of the peripheral immune system. In this review, we focus on the role of miRNAs in the regulation of BCR signaling as it pertains to B lymphocyte development and function. In particular, we discuss the most recent studies describing the role of miRNAs in the regulation of both early B cell development and peripheral B cell responses and examine the ways by which miRNAs regulate signal downstream of B cell antigen receptor to prevent aberrant activation and autoimmunity.
PMID: 34523719
ISSN: 1600-065x
CID: 5012342

Methotrexate hampers immunogenicity to BNT162b2 mRNA COVID-19 vaccine in immune-mediated inflammatory disease

Haberman, Rebecca H; Herati, Ramin; Simon, David; Samanovic, Marie; Blank, Rebecca B; Tuen, Michael; Koralov, Sergei; Atreya, Raja; Tascilar, Koray; Allen, Joseph; Castillo, Rochelle; Cornelius, Amber; Rackoff, Paula; Solomon, Gary; Adhikari, Samrachana; Azar, Natalie; Rosenthal, Pamela; Izmirly, Peter; Samuels, Jonathan; Golden, Brian; Reddy, Soumya M; Neurath, Markus; Abramson, Steven B; Schett, Georg; Mulligan, Mark; Scher, Jose U
PMID: 34035003
ISSN: 1468-2060
CID: 4888812

Staphylococcus Aureus Induces Stat5 Dependent Mir-155 Expression In Cutaneous T-Cell Lymphoma (Ctcl)

Willerslev-Olsen, Andreas; Rahbek Gjerdrum, Lise Mette; Lindahl, Lise M; Buus, Terkild B; Pallesen, Emil M H; Gluud, Maria; Bzorek, Michael; Nielsen, Boye S; Kamstrup, Maria R; Rittig, Anne Hald; Bonefeld, Charlotte M; Krejsgaard, Thorbjørn; Geisler, Carsten; Koralov, Sergei B; Litman, Thomas; Becker, Jurgen C; Woetmann, Anders; Iversen, Lars; Odum, Niels
Staphylococcus aureus enterotoxins (SE) are believed to fuel disease activity in cutaneous T-cell lymphoma (CTCL). Recent data support this by showing that antibiotics inhibit malignant T cells in skin lesions in mycosis fungoides and Sezary syndrome, the most common forms of CTCL. Yet, it remains incompletely characterized how SE fuel disease activity. Here, we show that SE induce expression of the oncogenic microRNA mir-155 in primary malignant T cells. Thus, SE and S. aureus-isolates from lesional patient skin induce mir-155 expression, at least partly, through the IL-2Rg/JAK/STAT5 pathway, and the effect is augmented by the presence of non-malignant T cells. Importantly, mycosis fungoides lesions harbor S. aureus, express pY-STAT5, and display enhanced mir-155 expression, when compared with non-lesional and healthy skin. Preliminary data show that aggressive antibiotic therapy is associated with decreased pY-STAT5 and mir-155 expression in lesional skin in two patients with Sezary syndrome. In conclusion, we demonstrate that S. aureus and its enterotoxins induce enhanced expression of oncogenic mir-155 providing mechanistic insight into the role of S. aureus in CTCL. Our findings support that environmental stimuli such as bacteria can fuel disease progression in CTCL.
PMID: 33862068
ISSN: 1523-1747
CID: 4846412

Methotrexate hampers immunogenicity to BNT162B2 mRNA COVID-19 vaccine in immune-mediated inflammatory disease [Meeting Abstract]

Haberman, R; Herati, R; Simon, D; Samanovic, M; Tuen, M; Blank, R; Koralov, S; Atreya, R; Tascilar, K; Allen, J; Castillo, R; Cornelius, A; Rackoff, P; Solomon, G; Adhikari, S; Azar, N; Rosenthal, P; Izmirly, P; Samuels, J; Golden, B; Reddy, S; Neurath, M; Abramson, S B; Schett, G; Mulligan, M; Scher, J
Background/Purpose: Patients with immune mediated inflammatory disorders (IMIDs) have an inherently heightened susceptibility to infection and may be considered high risk for developing COVID-19. While data regarding the COVID-19 vaccine's immunogenicity in an immunocompetent adult population is rapidly emerging, the ability of IMID patients to adequately respond to these vaccines is not known. Here, we investigate the humoral and cellular immune response to mRNA COVID-19 vaccines in patients with IMIDs on immunomodulatory treatment Methods: Patients with immune mediated inflammatory disorders (IMIDs) have an inherently heightened susceptibility to infection and may be considered high risk for developing COVID-19. While data regarding the COVID-19 vaccine's immunogenicity in an immunocompetent adult population is rapidly emerging, the ability of IMID patients to adequately respond to these vaccines is not known. Here, we investigate the humoral and cellular immune response to mRNA COVID-19 vaccines in patients with IMIDs on immunomodulatory treatment.
Result(s): The NY cohort baseline characteristics are found in Table 1. The Erlangen cohort consisted of 182 healthy subjects, 11 subjects with IMID receiving TNFi monotherapy, and 20 subjects with IMID on MTX monotherapy. In both cohorts, healthy individuals and those with IMID not on MTX were similar in age, while those IMID patients receiving MTX were generally older. In the NY cohort, of the healthy participants, 96.3% demonstrated adequate humoral immune response. Patients with IMID not on MTX achieved a similar rate of high antibody response rate (91.8%), while those on MTX had a lower rate of adequate humoral response (75.0%) (Figure 1A). This remains true even after the exclusion of patients who had evidence of prior COVID-19 infection (P= 0.014). Of note, 3 out of the 4 IMID patients receiving rituximab did not produce an adequate response. Similarly, in the Erlangen validation cohort, 98.3% of healthy controls, 90.9% of patients with IMID receiving TNFi monotherapy, and 50.0% receiving MTX monotherapy achieved adequate immunogenicity (Figure 1B). These differences remain significant when combining the cohorts, using a stricter definition of adequate response, and in a subgroup analysis by age. Cellular response was also analyzed in a subgroup of the NY cohort before and after second vaccination. Activated CD8+ T cells (CD8+ T cells expressing Ki67 and CD38) and the granzyme B-producing subset of these activated CD8+ T cells, were induced in immunocompetent adults and those with IMID not on MTX, but not induced in patients receiving MTX (Figure 2).
Conclusion(s): In two independent cohorts of IMID patients, MTX, a widely used immunomodulator for the treatment of several IMIDs, adversely affected humoral and cellular immune response to COVID-19 mRNA vaccines. Although precise cut offs for immunogenicity that correlate with vaccine efficacy are yet to be established, our findings suggest that different strategies may need to be explored in patients with IMID taking MTX to increase the chances of immunization efficacy against SARS-CoV-2, as has been demonstrated for other viral vaccines
PMCID:
EMBASE:637275567
ISSN: 2326-5205
CID: 5164692

Poor antigen-specific responses to the second BNT162b2 mRNA vaccine dose in SARS-CoV-2-experienced individuals

Samanovic, Marie I; Cornelius, Amber R; Wilson, Jimmy P; Karmacharya, Trishala; Gray-Gaillard, Sophie L; Allen, Joseph Richard; Hyman, Sara Wesley; Moritz, Gali; Ali, Mahnoor; Koralov, Sergei B; Mulligan, Mark J; Herati, Ramin Sedaghat
The advent of COVID-19 vaccines will play a major role in helping to end the pandemic that has killed millions worldwide. Vaccine candidates have demonstrated robust humoral responses and have protected against infection. However, efficacy trials were focused on individuals with no prior exposure to SARS-CoV-2, and, as a result, little is known about immune responses induced by these mRNA vaccines in individuals who recovered from COVID-19. Here, we evaluated immune responses in 32 subjects who received two-dose BNT162b2 mRNA vaccination. In individuals naive to SARS-CoV-2, we observed robust increases in humoral and antigen-specific antibody-secreting cell (ASC) responses following each dose of vaccine, whereas individuals with prior exposure to SARS-CoV-2 demonstrated strong humoral and antigen-specific ASC responses to the first dose but muted responses to the second dose of the vaccine for the time points studied. These data highlight an important gap in our knowledge and may have major implications for how these vaccines should be used to prevent COVID-19.
PMCID:7885942
PMID: 33594383
ISSN: n/a
CID: 4786862