37 results on '"Beata Majchrzak-Kita"'
Search Results
2. Prospective observational study and serosurvey of SARS-CoV-2 infection in asymptomatic healthcare workers at a Canadian tertiary care center.
- Author
-
Victor H Ferreira, Andrzej Chruscinski, Vathany Kulasingam, Trevor J Pugh, Tamara Dus, Brad Wouters, Amit Oza, Matthew Ierullo, Terrance Ku, Beata Majchrzak-Kita, Sonika T Humar, Ilona Bahinskaya, Natalia Pinzon, Jianhua Zhang, Lawrence E Heisler, Paul M Krzyzanowski, Bernard Lam, Ilinca M Lungu, Dorin Manase, Krista M Pace, Pouria Mashouri, Michael Brudno, Michael Garrels, Tony Mazzulli, Myron Cybulsky, Atul Humar, and Deepali Kumar
- Subjects
Medicine ,Science - Abstract
Health care workers (HCWs) are at higher risk for SARS-CoV-2 infection and may play a role in transmitting the infection to vulnerable patients and members of the community. This is particularly worrisome in the context of asymptomatic infection. We performed a cross-sectional study looking at asymptomatic SARS-CoV-2 infection in HCWs. We screened asymptomatic HCWs for SARS-CoV-2 via PCR. Complementary viral genome sequencing was performed on positive swab specimens. A seroprevalence analysis was also performed using multiple assays. Asymptomatic health care worker cohorts had a combined swab positivity rate of 29/5776 (0.50%, 95%CI 0.32-0.75) relative to a comparative cohort of symptomatic HCWs, where 54/1597 (3.4%) tested positive for SARS-CoV-2 (ratio of symptomatic to asymptomatic 6.8:1). SARS-CoV-2 seroprevalence among 996 asymptomatic HCWs with no prior known exposure to SARS-CoV-2 was 1.4-3.4%, depending on assay. A novel in-house Coronavirus protein microarray showed differing SARS-CoV-2 protein reactivities and helped define likely true positives vs. suspected false positives. Our study demonstrates the utility of routine screening of asymptomatic HCWs, which may help to identify a significant proportion of infections.
- Published
- 2021
- Full Text
- View/download PDF
3. Central Role of ULK1 in Type I Interferon Signaling
- Author
-
Diana Saleiro, Swarna Mehrotra, Barbara Kroczynska, Elspeth M. Beauchamp, Pawel Lisowski, Beata Majchrzak-Kita, Tushar D. Bhagat, Brady L. Stein, Brandon McMahon, Jessica K. Altman, Ewa M. Kosciuczuk, Darren P. Baker, Chunfa Jie, Nadereh Jafari, Craig B. Thompson, Ross L. Levine, Eleanor N. Fish, Amit K. Verma, and Leonidas C. Platanias
- Subjects
Biology (General) ,QH301-705.5 - Abstract
We provide evidence that the Unc-51-like kinase 1 (ULK1) is activated during engagement of the type I interferon (IFN) receptor (IFNR). Our studies demonstrate that the function of ULK1 is required for gene transcription mediated via IFN-stimulated response elements (ISRE) and IFNγ activation site (GAS) elements and controls expression of key IFN-stimulated genes (ISGs). We identify ULK1 as an upstream regulator of p38α mitogen-activated protein kinase (MAPK) and establish that the regulatory effects of ULK1 on ISG expression are mediated possibly by engagement of the p38 MAPK pathway. Importantly, we demonstrate that ULK1 is essential for antiproliferative responses and type I IFN-induced antineoplastic effects against malignant erythroid precursors from patients with myeloproliferative neoplasms. Together, these data reveal a role for ULK1 as a key mediator of type I IFNR-generated signals that control gene transcription and induction of antineoplastic responses.
- Published
- 2015
- Full Text
- View/download PDF
4. A Rapid Screening Assay Identifies Monotherapy with Interferon-ß and Combination Therapies with Nucleoside Analogs as Effective Inhibitors of Ebola Virus.
- Author
-
Stephen D S McCarthy, Beata Majchrzak-Kita, Trina Racine, Hannah N Kozlowski, Darren P Baker, Thomas Hoenen, Gary P Kobinger, Eleanor N Fish, and Donald R Branch
- Subjects
Arctic medicine. Tropical medicine ,RC955-962 ,Public aspects of medicine ,RA1-1270 - Abstract
To date there are no approved antiviral drugs for the treatment of Ebola virus disease (EVD). While a number of candidate drugs have shown limited efficacy in vitro and/or in non-human primate studies, differences in experimental methodologies make it difficult to compare their therapeutic effectiveness. Using an in vitro model of Ebola Zaire replication with transcription-competent virus like particles (trVLPs), requiring only level 2 biosafety containment, we compared the activities of the type I interferons (IFNs) IFN-α and IFN-ß, a panel of viral polymerase inhibitors (lamivudine (3TC), zidovudine (AZT) tenofovir (TFV), favipiravir (FPV), the active metabolite of brincidofovir, cidofovir (CDF)), and the estrogen receptor modulator, toremifene (TOR), in inhibiting viral replication in dose-response and time course studies. We also tested 28 two- and 56 three-drug combinations against Ebola replication. IFN-α and IFN-ß inhibited viral replication 24 hours post-infection (IC50 0.038μM and 0.016μM, respectively). 3TC, AZT and TFV inhibited Ebola replication when used alone (50-62%) or in combination (87%). They exhibited lower IC50 (0.98-6.2μM) compared with FPV (36.8μM), when administered 24 hours post-infection. Unexpectedly, CDF had a narrow therapeutic window (6.25-25μM). When dosed >50μM, CDF treatment enhanced viral infection. IFN-ß exhibited strong synergy with 3TC (97.3% inhibition) or in triple combination with 3TC and AZT (95.8% inhibition). This study demonstrates that IFNs and viral polymerase inhibitors may have utility in EVD. We identified several 2 and 3 drug combinations with strong anti-Ebola activity, confirmed in studies using fully infectious ZEBOV, providing a rationale for testing combination therapies in animal models of lethal Ebola challenge. These studies open up new possibilities for novel therapeutic options, in particular combination therapies, which could prevent and treat Ebola infection and potentially reduce drug resistance.
- Published
- 2016
- Full Text
- View/download PDF
5. Neutralization against Omicron variant in transplant recipients after three doses of mRNA vaccine
- Author
-
Deepali Kumar, Queenie Hu, Reuben Samson, Victor H. Ferreira, Victoria G. Hall, Matthew Ierullo, Beata Majchrzak-Kita, William Hardy, Anne-Claude Gingras, and Atul Humar
- Subjects
Transplantation ,Neutralization Tests ,SARS-CoV-2 ,Spike Glycoprotein, Coronavirus ,COVID-19 ,Humans ,Immunology and Allergy ,Pharmacology (medical) ,Antibodies, Viral ,Antibodies, Neutralizing ,Transplant Recipients ,2019-nCoV Vaccine mRNA-1273 - Abstract
The SARS-CoV-2 virus Omicron variant has now supplanted wild-type virus as the dominant circulating strain globally. Three doses of mRNA COVID-19 vaccine are recommended for transplant recipients as their primary vaccine series. However, the immunogenicity of mRNA vaccines as they specifically relate to the Omicron variant are not well studied. We analyzed Omicron-specific neutralization in transplant recipients after three-doses of mRNA-1273 vaccine. Neutralization was determined using a SARS-CoV-2 spike pseudotyped lentivirus assay with constructs for Omicron and Delta variants. A total of 60 transplant patients (kidney, kidney-pancreas, lung, heart, liver) were analyzed 1 month and 3 months after completion of three doses of mRNA-1273. At 1 month, 11/60 (18.3%) patients had detectable neutralizing antibody responses to Omicron (log
- Published
- 2022
- Full Text
- View/download PDF
6. Neutralization of SARS-CoV-2 Variants in Transplant Recipients After Two and Three Doses of mRNA-1273 Vaccine
- Author
-
Deepali Kumar, Victoria G Hall, Anne-Claude Gingras, Matthew Ierullo, George Tomlinson, Atul Humar, Queenie Hu, Beata Majchrzak-Kita, Terrance Ku, Victor H Ferreira, and Reuben Samson
- Subjects
Male ,medicine.medical_specialty ,Alpha (ethology) ,Placebo ,Virus ,Neutralization ,Organ transplantation ,law.invention ,Immunocompromised Host ,Double-Blind Method ,Randomized controlled trial ,law ,Internal Medicine ,Humans ,Medicine ,skin and connective tissue diseases ,Neutralizing antibody ,Aged ,Original Research ,biology ,SARS-CoV-2 ,business.industry ,fungi ,COVID-19 ,Organ Transplantation ,General Medicine ,Middle Aged ,biology.organism_classification ,Antibodies, Neutralizing ,Transplant Recipients ,body regions ,Lentivirus ,Immunology ,biology.protein ,Female ,business ,2019-nCoV Vaccine mRNA-1273 - Abstract
SARS-CoV-2 infection has been associated with increased morbidity and mortality in organ transplant recipients, and data are limited on 2-dose or 3-dose vaccine immunogenicity against SARS-CoV-2 variants in this population. In this secondary analysis of a randomized trial of a third dose of mRNA-1273 vaccine versus placebo, the authors assessed neutralizing antibody responses against SARS-CoV-2 variants in transplant recipients after 2 and 3 vaccine doses., Visual Abstract. SARS-CoV-2 Variant Neutralization After Vaccine in Transplant Recipients. SARS-CoV-2 infection has been associated with increased morbidity and mortality in organ transplant recipients, and data are limited on 2-dose or 3-dose vaccine immunogenicity against SARS-CoV-2 variants in this population. In this secondary analysis of a randomized trial of a third dose of mRNA-1273 vaccine versus placebo, the authors assessed neutralizing antibody responses against SARS-CoV-2 variants in transplant recipients after 2 and 3 vaccine doses. Visual Abstract. SARS-CoV-2 Variant Neutralization After Vaccine in Transplant Recipients. SARS-CoV-2 infection has been associated with increased morbidity and mortality in organ transplant recipients, and data are limited on 2-dose or 3-dose vaccine immunogenicity against SARS-CoV-2 variants in this population. In this secondary analysis of a randomized trial of a third dose of mRNA-1273 vaccine versus placebo, the authors assessed neutralizing antibody responses against SARS-CoV-2 variants in transplant recipients after 2 and 3 vaccine doses., Background: COVID-19 is more severe in transplant recipients. Variants of concern have supplanted wild-type virus. In transplant recipients, data are limited on 2-dose or 3-dose vaccine immunogenicity against variant viruses. Objective: To assess neutralizing antibody responses against SARS-CoV-2 variants in transplant recipients after 2 and 3 vaccine doses. Design: Secondary analysis of a randomized, double-blind, controlled trial of a third dose of mRNA-1273 vaccine versus placebo. (ClinicalTrials.gov: NCT04885907) Setting: Single-center transplant program. Patients: Organ transplant recipients. Intervention: Third dose of mRNA-1273 vaccine versus placebo. Measurements: Sera were analyzed for neutralization against wild-type virus and the Alpha, Beta, and Delta variants using a surrogate virus neutralization assay and a spike-pseudotyped lentivirus assay. Results: A total of 117 transplant recipients were analyzed (60 in the mRNA-1273 group and 57 in the placebo group). Sera were obtained before and 4 to 6 weeks after the third dose. After 2 doses, the proportion of patients with positive neutralization for all 3 variants was small compared with wild-type virus. After the third dose of mRNA-1273 vaccine, the proportion with a positive neutralization response versus placebo was improved for all 3 variants as measured by both assays. Based on the pseudovirus neutralization assay against the Delta variant, 33 of 60 (55%) patients were positive in the mRNA-1273 group versus 10 of 57 (18%) in the placebo group (difference, 37 [95% CI, 19 to 53] percentage points). The differences were 36 (CI, 17 to 51) percentage points for the Alpha variant and 31 (CI, 15 to 46) percentage points for the Beta variant. In the mRNA-1273 group, lower neutralization values were observed for variants compared with wild-type virus, especially the Beta variant. Limitations: There is no clear correlate of protection for neutralizing antibody. This was a secondary analysis. Conclusion: In organ transplant recipients, a third dose of mRNA vaccine increases neutralizing antibody response against SARS-CoV-2 variants compared with placebo. Primary Funding Source: Ajmera Transplant Centre.
- Published
- 2022
- Full Text
- View/download PDF
7. Prospective Clinical, Virologic, and Immunologic Assessment of COVID-19 in Transplant Recipients
- Author
-
Les Lilly, Aman Sidhu, S. Joseph Kim, Coleman Rotstein, Tina Marinelli, Matthew Ierullo, Deepali Kumar, Terrance Ku, Beata Majchrzak-Kita, Seyed M Hosseini-Moghaddam, Vathany Kulasingam, Michael McDonald, Shahid Husain, Jeffrey Schiff, Victor H Ferreira, and Atul Humar
- Subjects
Adult ,Male ,medicine.medical_specialty ,medicine.medical_treatment ,Population ,Disease ,Antibodies, Viral ,Severity of Illness Index ,Covid ,Immune system ,Internal medicine ,Pandemic ,medicine ,Humans ,Prospective Studies ,Viral shedding ,education ,Aged ,Transplantation ,education.field_of_study ,biology ,SARS-CoV-2 ,business.industry ,COVID-19 ,Immunosuppression ,Organ Transplantation ,Middle Aged ,Viral Load ,Transplant Recipients ,Virus Shedding ,ComputingMethodologies_DOCUMENTANDTEXTPROCESSING ,biology.protein ,Female ,Antibody ,business ,Viral load - Abstract
Supplemental Digital Content is available in the text., Background. Several studies have described the clinical features of COVID-19 in solid-organ transplant recipients. However, many have been retrospective or limited to more severe cases (hospitalized) and have not routinely included serial virological sampling (especially in outpatients) and immunologic assessment. Methods. Transplant patients diagnosed with COVID-19 based on a respiratory sample PCR were prospectively followed up to 90 d. Patients provided consent for convalescent serum samples and serial nasopharyngeal swabs for SARS-CoV-2 antibody (antinucleoprotein and anti-RBD) and viral load, respectively. Results. In the 161 SOT recipients diagnosed with COVID-19, the spectrum of disease ranged from asymptomatic infection (4.3%) to hospitalization (60.6%), supplemental oxygen requirement (43.1%), mechanical ventilation (22.7%), and death (15.6%). Increasing age (OR, 1.031; 95% CI, 1.001-1.062; P = 0.046) and ≥2 comorbid conditions (OR, 3.690; 95% CI, 1.418-9.615; P = 0.007) were associated with the need for supplemental oxygen. Allograft rejection was uncommon (3.7%) despite immunosuppression modification. Antibody response at ≥14 d postsymptoms onset was present in 90% (anti-RBD) and 76.7% (anti-NP) with waning of anti-NP titers and stability of anti-RBD over time. Median duration of nasopharyngeal positivity was 10.0 d (IQR, 5.5–18.0) and shedding beyond 30 d was observed in 6.7% of patients. The development of antibody did not have an impact on viral shedding. Conclusions. This study demonstrates the spectrum of COVID-19 illness in transplant patients. Risk factors for severe disease are identified. The majority form antibody by 2 wk with differential stability over time. Prolonged viral shedding was observed in a minority of patients. Reduction of immunosuppression was a safe strategy.
- Published
- 2021
- Full Text
- View/download PDF
8. Randomized Trial of a Third Dose of mRNA-1273 Vaccine in Transplant Recipients
- Author
-
Jeffrey Schiff, Victor H Ferreira, Deepali Kumar, C. Chaparro, Vathany Kulasingam, Nazia Selzner, Matthew Ierullo, Michael McDonald, Victoria G Hall, Terrance Ku, Beata Majchrzak-Kita, Atul Humar, and George Tomlinson
- Subjects
medicine.medical_specialty ,Messenger RNA ,biology ,Coronavirus disease 2019 (COVID-19) ,business.industry ,Immunogenicity ,General Medicine ,Organ transplantation ,law.invention ,surgical procedures, operative ,Immune system ,Immunization ,Randomized controlled trial ,law ,Correspondence ,Immunology ,biology.protein ,Medicine ,Antibody ,business - Abstract
A Third Vaccine Dose in Organ-Transplant Recipients It is known that people receiving immune suppressive therapy, such as recipients of solid-organ transplants, have a suboptimal response to SARS-C...
- Published
- 2021
- Full Text
- View/download PDF
9. Delayed-interval BNT162b2 mRNA COVID-19 vaccination enhances humoral immunity and induces robust T cell responses
- Author
-
Victoria G. Hall, Victor H. Ferreira, Heidi Wood, Matthew Ierullo, Beata Majchrzak-Kita, Kathy Manguiat, Alyssia Robinson, Vathany Kulasingam, Atul Humar, and Deepali Kumar
- Subjects
Adult ,CD4-Positive T-Lymphocytes ,Male ,SARS-CoV-2 ,Immunology ,Vaccination ,Immunization, Secondary ,COVID-19 ,CD8-Positive T-Lymphocytes ,Middle Aged ,Immunity, Humoral ,Interferon-gamma ,Immunology and Allergy ,Humans ,Interleukin-2 ,Female ,Vaccination Hesitancy ,BNT162 Vaccine ,Cells, Cultured - Abstract
Delayed dosing intervals are a strategy to immunize a greater proportion of the population. In an observational study, we compared humoral and cellular responses in health care workers receiving two doses of BNT162b2 (Pfizer-BioNTech) vaccine at standard (3- to 6-week) and delayed (8- to 16-week) intervals. In the delayed-interval group, anti-receptor-binding domain antibody titers were significantly enhanced compared to the standard-interval group. The 50% plaque reduction neutralization test (PRNT50) and PRNT90 titers against wild-type (ancestral) severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and Alpha, Beta and Delta variants were higher in the delayed-interval group. Spike-specific polyfunctional CD4
- Published
- 2021
10. Safety and Immunogenicity After a Three-Dose SARS-CoV-2 Vaccine Schedule in Allogeneic Stem Cell Transplant Recipients
- Author
-
Muneyoshi Kimura, Victor H. Ferreira, Sagar Kothari, Ivan Pasic, Jonas I. Mattsson, Vathany Kulasingam, Atul Humar, Allison Mah, Jean-Sébastien Delisle, Matthew Ierullo, Beata Majchrzak-Kita, Deepali Kumar, and Seyed M. Hosseini-Moghaddam
- Subjects
CD4-Positive T-Lymphocytes ,Immunity, Cellular ,Transplantation ,COVID-19 Vaccines ,SARS-CoV-2 ,Hematopoietic Stem Cell Transplantation ,Immunization, Secondary ,COVID-19 ,Graft vs Host Disease ,Cell Biology ,Hematology ,CD8-Positive T-Lymphocytes ,Immunity, Humoral ,Immunogenicity, Vaccine ,Humans ,Interleukin-2 ,Molecular Medicine ,Immunology and Allergy ,Prospective Studies - Abstract
In allogeneic stem cell transplant (Allo-SCT) recipients, the cell-mediated and humoral immunogenicity of the 3-dose SARS-CoV-2 vaccination schedule has not been investigated in prospective studies. In a prospective cohort, we recruited 122 Allo-SCT recipients since August 2021, when Ontario began offering a 3-dose vaccine schedule for Allo-SCT recipients. We determined humoral and cell-mediated immunity and adverse effects of the 3-dose SARS-COV-2 vaccination schedule in Allo-SCT recipients. In immunogenicity analysis (n = 95), the median (interquartile range [IQR]) antibody titer against the receptor-binding domain (RBD) of the spike (S) protein after the third dose (10,358.0 U/mL [IQR = 673.9-31,753.0]) was significantly higher than that after the first (10.2 U/mL [IQR = 0.6-37.0]) and the second doses (125.6 U/mL [IQR = 2.8-1251.0]) (P.0001). The haploidentical donor status was an independent risk factor (adjusted odds ratio = 7.67, 95% confidence interval [CI], 1.86-31.60) for suboptimal antibody response (anti-RBD100 U/mL). S-specific CD4
- Published
- 2022
- Full Text
- View/download PDF
11. Delayed interval BNT162b2 mRNA COVID-19 vaccination provides robust immunity
- Author
-
Vathany Kulasingam, Atul Humar, Beata Majchrzak-Kita, Deepali Kumar, Matthew Ierullo, Victoria G Hall, Victor H Ferreira, and Terrance Ku
- Subjects
Vaccination ,Messenger RNA ,Coronavirus disease 2019 (COVID-19) ,Immunity ,Immunology ,Interval (graph theory) ,Biology - Abstract
Shortages of COVID-19 vaccines have results in delayed dosing intervals as a strategy to immunize a greater proportion of the population. The effect of this strategy on vaccine immunogenicity is not well studied. Humoral (anti-RBD levels and neutralization) and cellular immune responses were compared in health care workers receiving two doses of BNT162b2 (Pfizer-BioNTech) vaccines at standard (3-6 week) and delayed (8-12 week) intervals. In the delayed group, anti-RBD antibody titres were significantly enhanced compared to the standard interval group. Neutralizing antibody responses were excellent and comparable in both groups. A slight decrease in Spike-specific polyfunctional CD4+ T-cells expressing interferon-γ and IL-2 as well as monofunctional CD4+ T-cells was seen in the delayed group. Both polyfunctional and monofunctional CD8+ T-cell responses were comparable. Our data suggest that the strategy of delayed second dose mRNA vaccination is not overtly detrimental, and specifically may lead to an enhanced humoral immune response.
- Published
- 2021
- Full Text
- View/download PDF
12. Severe Acute Respiratory Syndrome Coronavirus 2 Infection Induces Greater T-Cell Responses Compared to Vaccination in Solid Organ Transplant Recipients
- Author
-
Victoria G Hall, Victor H Ferreira, Beata Majchrzak-Kita, Matthew Ierullo, Vathany Kulasingam, Deepali Kumar, Atul Humar, Terrance Ku, and Tina Marinelli
- Subjects
COVID-19 Vaccines ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,T cell ,T-Lymphocytes ,T cells ,Disease ,Major Articles and Brief Reports ,Immunity ,Major Article ,Immunology and Allergy ,Medicine ,Humans ,Immunity, Cellular ,business.industry ,SARS-CoV-2 ,Vaccination ,COVID-19 ,Organ Transplantation ,Transplant Recipients ,Transplantation ,Infectious Diseases ,medicine.anatomical_structure ,AcademicSubjects/MED00290 ,Immunology ,business ,Solid organ transplantation ,CD8 ,transplantation - Abstract
T-cell immunity associated with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection or vaccination in solid organ transplant recipients (SOTRs) is poorly understood. To address this, we measured T-cell responses in 50 SOTRs with prior SARS-CoV-2 infection. The majority of patients mounted SARS-CoV-2–specific CD4+ T-cell responses against spike (S), nucleocapsid, and membrane proteins; CD8+ T-cell responses were generated to a lesser extent. CD4+ T-cell responses correlated with antibody levels. Severity of disease and mycophenolate dose were moderately associated with lower proportions of antigen-specific T cells. Relative to nontransplant controls, SOTRs had perturbations in both total and antigen-specific T cells, including higher frequencies of total PD-1+ CD4+ T cells. Vaccinated SOTRs (n = 55) mounted significantly lower proportions of S-specific polyfunctional CD4+ T cells after 2 doses, relative to unvaccinated SOTRs with prior coronavirus disease 2019. Together, these results suggest that SOTRs generate robust T-cell responses following natural infection that correlate with disease severity but generate comparatively lower T-cell responses following mRNA vaccination., Solid organ transplant recipients mount antigen-specific T-cell responses after SARS-CoV-2 infection that correlate with antibodies and disease severity. Compared to natural infection, vaccine responses to 2 doses of mRNA vaccine result in comparably lower frequencies of antigen-specific CD4+ T cells.
- Published
- 2021
13. Humoral and cellular immune response and safety of two‐dose SARS‐CoV‐2 mRNA‐1273 vaccine in solid organ transplant recipients
- Author
-
Tina Marinelli, Anila Yousuf, Beata Majchrzak-Kita, Victoria G Hall, Vathany Kulasingam, Atul Humar, Matthew Ierullo, Terrance Ku, Deepali Kumar, and Victor H Ferreira
- Subjects
COVID-19 Vaccines ,T cell ,infectious disease ,clinical research/practice ,Antibodies, Viral ,Neutralization ,Immune system ,infection and infectious agents ‐ viral ,vaccine ,Immunology and Allergy ,Medicine ,Humans ,Pharmacology (medical) ,Neutralizing antibody ,Transplantation ,Immunity, Cellular ,biology ,business.industry ,SARS-CoV-2 ,Immunogenicity ,COVID-19 ,Original Articles ,Organ Transplantation ,Antibodies, Neutralizing ,Vaccination ,medicine.anatomical_structure ,Immunology ,biology.protein ,Original Article ,Antibody ,business ,CD8 - Abstract
Solid organ transplant recipients are at high risk of severe disease from COVID-19. We assessed the immunogenicity of mRNA-1273 vaccine using a combination of antibody testing, surrogate neutralization assays, and T cell assays. Patients were immunized with two doses of vaccine and immunogenicity assessed after each dose using the above tests. CD4+ and CD8+ T cell responses were assessed in a subset using flow-cytometry. A total of 127 patients were enrolled of which 110 provided serum at all time points. A positive anti-RBD antibody was seen in 5.0% after one dose and 34.5% after two doses. Neutralizing antibody was present in 26.9%. Of note, 28.5% of patients with anti-RBD did not have neutralizing antibody. T cell responses in a sub-cohort of 48 patients showed a positive CD4+ T cell response in 47.9%. Of note, in this sub-cohort, 46.2% of patients with a negative anti-RBD, still had a positive CD4+ T cell response. The vaccine was safe and well-tolerated. In summary, immunogenicity of mRNA-1273 COVID-19 vaccine was modest, but a subset of patients still develop neutralizing antibody and CD4+T- cell responses. Importantly polyfunctional CD4+T cell responses were observed in a significant portion who were antibody negative, further highlighting the importance of vaccination in this patient population. IRB Statement: This study was approved by the University Health Network Research Ethics Board (CAPCR ID 20-6069).
- Published
- 2021
14. Prospective observational study and serosurvey of SARS-CoV-2 infection in asymptomatic healthcare workers at a Canadian tertiary care center
- Author
-
Vathany Kulasingam, Victor H Ferreira, Michael Brudno, Atul Humar, Bernard Lam, Myron I. Cybulsky, Trevor J. Pugh, Krista M. Pace, Natalia Pinzon, Matthew Ierullo, Ilinca Lungu, Brad Wouters, Sonika T. Humar, Paul M. Krzyzanowski, Tamara Dus, Michael Garrels, Amit M. Oza, Beata Majchrzak-Kita, Andrzej Chruscinski, Ilona Bahinskaya, Pouria Mashouri, Deepali Kumar, Tony Mazzulli, Terrance Ku, Dorin Manase, Jianhua Zhang, and Lawrence E. Heisler
- Subjects
RNA viruses ,Viral Diseases ,Pulmonology ,Coronaviruses ,Microarrays ,Physiology ,viruses ,Biochemistry ,Serology ,Tertiary Care Centers ,Medical Conditions ,Seroepidemiologic Studies ,Immune Physiology ,Epidemiology ,Health care ,Medical Personnel ,Enzyme-Linked Immunoassays ,Asymptomatic Infections ,Pathology and laboratory medicine ,Virus Testing ,Immune System Proteins ,Multidisciplinary ,virus diseases ,Medical microbiology ,Professions ,Infectious Diseases ,Bioassays and Physiological Analysis ,COVID-19 Nucleic Acid Testing ,Viruses ,Cohort ,Medicine ,SARS CoV 2 ,Pathogens ,medicine.symptom ,Research Article ,Canada ,medicine.medical_specialty ,SARS coronavirus ,Health Personnel ,Science ,Immunology ,Context (language use) ,Research and Analysis Methods ,Microbiology ,Asymptomatic ,Antibodies ,COVID-19 Serological Testing ,Respiratory Disorders ,Diagnostic Medicine ,Internal medicine ,medicine ,Humans ,Seroprevalence ,Immunoassays ,Medicine and health sciences ,Biology and life sciences ,business.industry ,Organisms ,Viral pathogens ,Proteins ,COVID-19 ,Covid 19 ,Microbial pathogens ,Respiratory Infections ,People and Places ,Immunologic Techniques ,Population Groupings ,business - Abstract
Health care workers (HCWs) are at higher risk for SARS-CoV-2 infection and may play a role in transmitting the infection to vulnerable patients and members of the community. This is particularly worrisome in the context of asymptomatic infection. We performed a cross-sectional study looking at asymptomatic SARS-CoV-2 infection in HCWs. We screened asymptomatic HCWs for SARS-CoV-2 via PCR. Complementary viral genome sequencing was performed on positive swab specimens. A seroprevalence analysis was also performed using multiple assays. Asymptomatic health care worker cohorts had a combined swab positivity rate of 29/5776 (0.50%, 95%CI 0.32–0.75) relative to a comparative cohort of symptomatic HCWs, where 54/1597 (3.4%) tested positive for SARS-CoV-2 (ratio of symptomatic to asymptomatic 6.8:1). SARS-CoV-2 seroprevalence among 996 asymptomatic HCWs with no prior known exposure to SARS-CoV-2 was 1.4–3.4%, depending on assay. A novel in-house Coronavirus protein microarray showed differing SARS-CoV-2 protein reactivities and helped define likely true positives vs. suspected false positives. Our study demonstrates the utility of routine screening of asymptomatic HCWs, which may help to identify a significant proportion of infections.
- Published
- 2021
15. Prospective Observational Study of Screening Asymptomatic Healthcare Workers for SARS-CoV-2 at a Canadian Tertiary Care Center
- Author
-
Brad Wouters, Michael Garrels, Myron I. Cybulsky, Victor H Ferreira, Krista M. Pace, Pouria Mashouri, Matthew Ierullo, Michael Brudno, Deepali Kumar, Andrzej Chruscinski, Atul Humar, Bernard Lam, Beata Majchrzak-Kita, Ilinca Lungu, Tony Mazzulli, Tamara Dus, Amit M. Oza, Trevor J. Pugh, Natalia Pinzon, Ilona Bahinskaya, Lawrence E. Heisler, Dorin Manase, Vathany Kulasingam, Sonika T. Humar, Paul M. Krzyzanowski, Terrance J.Y. Ku, and Jianhua Zhang
- Subjects
medicine.medical_specialty ,business.industry ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Tertiary care ,Asymptomatic ,Internal medicine ,Health care ,Cohort ,medicine ,False positive paradox ,Seroprevalence ,Observational study ,medicine.symptom ,business - Abstract
We screened three separate cohorts of healthcare workers for SARS-CoV-2 via nasopharyngeal swab PCR. A seroprevalence analysis using multiple assays was performed in a subgroup. The asymptomatic health care worker cohorts had a combined swap positivity rate of 29/5776 (0.50%, 95%CI 0.32-0.75) compared to the symptomatic cohort rate of 54/1597 (3.4%) (ratio of symptomatic to asymptomatic 6.8:1). Sequencing demonstrated several variants. The seroprevalence (n=996) was 1.4-3.4% depending on assay. Protein microarray analysis showed differing SARS-CoV-2 protein reactivities and helped define likely true positives vs. suspected false positives. Routine screening of asymptomatic health care workers helps identify a significant proportion of infections.
- Published
- 2020
- Full Text
- View/download PDF
16. Small Molecule Agonists for the Type I Interferon Receptor: An In Silico Approach
- Author
-
Melissa Maureen Lewis, Beata Majchrzak-Kita, Noruê Salum, Angelica M. Bello, Eleanor N. Fish, Lakshmi P. Kotra, and Lianhu Wei
- Subjects
Models, Molecular ,0301 basic medicine ,Peptidomimetic ,In silico ,Immunology ,Gene Expression ,Alpha interferon ,Cold storage ,Receptor, Interferon alpha-beta ,Pharmacology ,Biology ,Ligands ,Antiviral Agents ,Protein Structure, Secondary ,Small Molecule Libraries ,Structure-Activity Relationship ,User-Computer Interface ,03 medical and health sciences ,Cell Line, Tumor ,Virology ,Humans ,Structure–activity relationship ,Computer Simulation ,Encephalomyocarditis virus ,Receptor ,B-Lymphocytes ,Interferon-alpha ,Cell Biology ,Small molecule ,Recombinant Proteins ,High-Throughput Screening Assays ,030104 developmental biology ,Drug Design ,Peptidomimetics - Abstract
Type I interferons (IFNs) exhibit broad-spectrum antiviral activity, with potential utility against emerging acute virus infections that pose a threat to global health. Recombinant IFN-αs that have been approved for clinical use require cold storage and are administered through intramuscular or subcutaneous injection, features that are problematic for global distribution, storage, and administration. Cognizant that the biological potency of an IFN-α subtype is determined by its binding affinity to the type I IFN receptor, IFNAR, we identified a panel of small molecule nonpeptide compounds using an in silico screening strategy that incorporated specific structural features of amino acids in the receptor-binding domains of the most potent IFN-α, IFN alfacon-1. Hit compounds were selected based on ease of synthesis and formulation properties. In preliminary biological assays, we provide evidence that these compounds exhibit antiviral activity. This proof-of-concept study validates the strategy of in silico design and development for IFN mimetics.
- Published
- 2016
- Full Text
- View/download PDF
17. Sirtuin 2–mediated deacetylation of cyclin-dependent kinase 9 promotes STAT1 signaling in type I interferon responses
- Author
-
Caroline Driehaus, Barbara Kroczynska, Anna Rogalska, Leonidas C. Platanias, David Gius, Thomas Lienhoop, Swarna Mehrotra, Diana Saleiro, Athanassios Vassilopoulos, Ewa M. Kosciuczuk, Beata Majchrzak-Kita, Eleanor N. Fish, Pawel Lisowski, and Acara Turner
- Subjects
0301 basic medicine ,Transcription, Genetic ,SIRT2 ,Biochemistry ,03 medical and health sciences ,Mice ,Sirtuin 2 ,Interferon ,medicine ,Animals ,Humans ,STAT1 ,Phosphorylation ,Molecular Biology ,Mice, Knockout ,030102 biochemistry & molecular biology ,biology ,Kinase ,Acetylation ,Cell Biology ,U937 Cells ,Cyclin-Dependent Kinase 9 ,Cell biology ,030104 developmental biology ,STAT1 Transcription Factor ,Sirtuin ,Interferon Type I ,biology.protein ,Cyclin-dependent kinase 9 ,Signal transduction ,Janus kinase ,medicine.drug ,Signal Transduction - Abstract
Type I interferons (IFNs) induce expression of multiple genes that control innate immune responses to invoke both antiviral and antineoplastic activities. Transcription of these interferon-stimulated genes (ISGs) occurs upon activation of the canonical Janus kinase (JAK)–signal transducer and activator of transcription (STAT) signaling pathways. Phosphorylation and acetylation are both events crucial to tightly regulate expression of ISGs. Here, using mouse embryonic fibroblasts and an array of biochemical methods including immunoblotting and kinase assays, we show that sirtuin 2 (SIRT2), a member of the NAD-dependent protein deacetylase family, is involved in type I IFN signaling. We found that SIRT2 deacetylates cyclin-dependent kinase 9 (CDK9) in a type I IFN–dependent manner and that the CDK9 deacetylation is essential for STAT1 phosphorylation at Ser-727. We also found that SIRT2 is subsequently required for the transcription of ISGs and for IFN-driven antiproliferative responses in both normal and malignant cells. These findings establish the existence of a previously unreported signaling pathway whose function is essential for the control of JAK–STAT signaling and the regulation of IFN responses. Our findings suggest that targeting sirtuin activities may offer an avenue in the development of therapies for managing immune-related diseases and cancer.
- Published
- 2018
18. IFN-γ-inducible antiviral responses require ULK1-mediated activation of MLK3 and ERK5
- Author
-
Mariafausta Fischietti, Eleanor N. Fish, Patrick A. Ozark, Gavin T. Blyth, Leonidas C. Platanias, Neha K. Reddy, Diana Saleiro, Roger J. Davis, Curt M. Horvath, Ewa M. Kosciuczuk, Beata Majchrzak-Kita, and Ahmet Dirim Arslan
- Subjects
0301 basic medicine ,Transcription, Genetic ,Biochemistry ,Article ,03 medical and health sciences ,Interferon-gamma ,Mice ,0302 clinical medicine ,Interferon ,Cell Line, Tumor ,medicine ,Autophagy ,Animals ,Autophagy-Related Protein-1 Homolog ,Humans ,STAT1 ,Phosphorylation ,Protein kinase A ,Molecular Biology ,Transcription factor ,Mitogen-Activated Protein Kinase 7 ,Receptors, Interferon ,biology ,Kinase ,Chemistry ,Gene Expression Profiling ,Intracellular Signaling Peptides and Proteins ,Cell Biology ,U937 Cells ,ULK1 ,MAP Kinase Kinase Kinases ,Class III Phosphatidylinositol 3-Kinases ,Immunity, Innate ,Recombinant Proteins ,Cell biology ,030104 developmental biology ,Virus Diseases ,Multigene Family ,biology.protein ,STAT protein ,Cytokines ,Beclin-1 ,Signal transduction ,030215 immunology ,medicine.drug ,Protein Binding ,Signal Transduction - Abstract
It is well established that activation of the transcription factor signal transducer and activator of transcription 1 (STAT1) is required for the interferon-γ (IFN-γ)-mediated antiviral response. Here, we found that IFN-γ receptor stimulation also activated Unc-51-like kinase 1 (ULK1), an initiator of Beclin-1-mediated autophagy. Furthermore, the interaction between ULK1 and the mitogen-activated protein kinase kinase kinase MLK3 (mixed lineage kinase 3) was necessary for MLK3 phosphorylation and downstream activation of the kinase ERK5. This autophagy-independent activity of ULK1 promoted the transcription of key antiviral IFN-stimulated genes (ISGs) and was essential for IFN-γ-dependent antiviral effects. These findings define a previously unknown IFN-γ pathway that appears to be a key element of the antiviral response.
- Published
- 2018
19. Interferon γ (IFNγ) Signaling via Mechanistic Target of Rapamycin Complex 2 (mTORC2) and Regulatory Effects in the Generation of Type II Interferon Biological Responses
- Author
-
Ewa M. Kosciuczuk, Zofia Warminska, Eleanor N. Fish, Diana Saleiro, Barbara Kroczynska, Leonidas C. Platanias, Swarna Mehrotra, Gavin T. Blyth, Jacek Jemielity, Robert L. Rafidi, Ahmet Dirim Arslan, and Beata Majchrzak-Kita
- Subjects
0301 basic medicine ,Mechanistic Target of Rapamycin Complex 2 ,mTORC1 ,Mechanistic Target of Rapamycin Complex 1 ,Biology ,Antiviral Agents ,Biochemistry ,Interferon-gamma ,Mice ,03 medical and health sciences ,Eukaryotic initiation factor 4F ,Interferon ,medicine ,Animals ,Humans ,Interferon gamma ,Phosphorylation ,Molecular Biology ,Mechanistic target of rapamycin ,PI3K/AKT/mTOR pathway ,Receptors, Interferon ,Mice, Knockout ,Eukaryotic Translation Initiation Factor 4F ,TOR Serine-Threonine Kinases ,U937 Cells ,Cell Biology ,Fibroblasts ,Hematopoietic Stem Cells ,Interferon-Stimulated Gene Factor 3, gamma Subunit ,Hematopoiesis ,Cell biology ,Chemokine CXCL10 ,Rapamycin-Insensitive Companion of mTOR Protein ,030104 developmental biology ,Eukaryotic Initiation Factor-4F ,Gene Expression Regulation ,Multiprotein Complexes ,Polyribosomes ,Protein Biosynthesis ,Immunology ,biology.protein ,Signal transduction ,Carrier Proteins ,Signal Transduction ,medicine.drug - Abstract
We provide evidence for a unique pathway engaged by the type II IFN receptor, involving mTORC2/AKT-mediated downstream regulation of mTORC1 and effectors. These events are required for formation of the eukaryotic translation initiation factor 4F complex (eIF4F) and initiation of mRNA translation of type II interferon-stimulated genes. Our studies establish that Rictor is essential for the generation of type II IFN-dependent antiviral and antiproliferative responses and that it controls the generation of type II IFN-suppressive effects on normal and malignant hematopoiesis. Together, our findings establish a central role for mTORC2 in IFNγ signaling and type II IFN responses.
- Published
- 2016
- Full Text
- View/download PDF
20. Slfn2 Regulates Type I Interferon Responses by Modulating the NF-κB Pathway
- Author
-
Eleanor N. Fish, Ahmet Dirim Arslan, Mariafausta Fischietti, Antonella Sassano, Beata Majchrzak-Kita, Diana Saleiro, Hidayatullah G. Munshi, Kazumi Ebine, and Leonidas C. Platanias
- Subjects
0301 basic medicine ,Protein subunit ,Phosphatase ,Gene Expression ,Cell Cycle Proteins ,Biology ,Models, Biological ,Nuclear factor kappa b ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Transcription (biology) ,Interferon ,medicine ,Phosphoprotein Phosphatases ,Animals ,Humans ,Promoter Regions, Genetic ,Molecular Biology ,Gene ,Cells, Cultured ,Mice, Knockout ,Binding Sites ,NF-kappa B ,NF-κB ,Cell Biology ,Cell biology ,Mice, Inbred C57BL ,030104 developmental biology ,chemistry ,030220 oncology & carcinogenesis ,Interferon Type I ,NIH 3T3 Cells ,Signal transduction ,medicine.drug ,Signal Transduction ,Research Article - Abstract
Although members of the Slfn family have been implicated in the regulation of type I interferon (IFN) responses, the mechanisms by which they mediate their effects remain unknown. In the present study, we provide evidence that targeted disruption of the Slfn2 gene leads to increased transcription of IFN-stimulated genes (ISGs) and enhanced type I IFN-mediated antiviral responses. We demonstrate that Slfn2 interacts with protein phosphatase 6 regulatory subunit 1 (PPP6R1), leading to reduced type I IFN-induced activation of nuclear factor kappa B (NF-κB) signaling, resulting in reduced expression of ISGs. Altogether, these data suggest a novel mechanism by which Slfn2 controls ISG expression and provide evidence for a critical role for Slfn2 in the regulation of IFN-mediated biological responses.
- Published
- 2018
21. Small molecule mimetics of an interferon-α receptor interacting domain
- Author
-
Beata Majchrzak-Kita, Noruê Salum, Angelica M. Bello, Eleanor N. Fish, Meena K. Purohit, Lianhu Wei, and Lakshmi P. Kotra
- Subjects
Models, Molecular ,Protein Conformation ,Peptidomimetic ,Clinical Biochemistry ,Drug Evaluation, Preclinical ,Pharmaceutical Science ,Receptor, Interferon alpha-beta ,medicine.disease_cause ,Biochemistry ,Epitope ,Cell Line ,Small Molecule Libraries ,Epitopes ,Cell surface receptor ,Drug Discovery ,medicine ,Humans ,Phosphorylation ,Receptor ,Molecular Biology ,Aspartic Acid ,Chemistry ,Drug discovery ,Molecular Mimicry ,Organic Chemistry ,Interferon-alpha ,Small molecule ,Protein Structure, Tertiary ,Molecular mimicry ,STAT1 Transcription Factor ,Molecular Medicine ,Peptides - Abstract
Small molecules that mimic IFN-α epitopes that interact with the cell surface receptor, IFNAR, would be useful therapeutics. One such 8-amino acid region in IFN-α2, designated IRRP-1, was used to derive 11 chemical compounds that belong to 5 distinct chemotypes, containing the molecular features represented by the key residues Leu30, Arg33, and Asp35 in IRRP-1. Three of these compounds exhibited potential mimicry to IRRP-1 and, in cell based assays, as predicted, effectively inhibited IFNAR activation by IFN-α. Of these, compound 3 did not display cell toxicity and reduced IFN-α-inducible STAT1 phosphorylation and STAT-DNA binding. Based on physicochemical properties’ analyses, our data suggest that moieties with acidic p K a on the small molecule may be a necessary element for mimicking the carboxyl group of Asp35 in IRRP-1. Our data confirm the relevance of this strategy of molecular mimicry of ligand–receptor interaction domains of protein partners for small molecule drug discovery.
- Published
- 2014
- Full Text
- View/download PDF
22. Central Regulatory Role for SIN1 in Interferon γ (IFNγ) Signaling and Generation of Biological Responses*
- Author
-
Ewa M. Kosciuczuk, Leonidas C. Platanias, Bing Su, Lucy Xu, Beata Majchrzak-Kita, Eleanor N. Fish, Barbara Kroczynska, Diana Saleiro, Robert L. Rafidi, Elizabeth A. Eklund, Jacek Jemielity, Gavin T. Blyth, and Jessica K. Altman
- Subjects
0301 basic medicine ,mTORC1 ,Biology ,Biochemistry ,Cell Line ,03 medical and health sciences ,chemistry.chemical_compound ,Interferon-gamma ,Mice ,Animals ,Humans ,STAT1 ,Phosphorylation ,Molecular Biology ,Adaptor Proteins, Signal Transducing ,Eukaryotic Translation Initiation Factor 4F ,Translation (biology) ,Tyrosine phosphorylation ,Cell Biology ,Immunity, Innate ,Cell biology ,030104 developmental biology ,STAT1 Transcription Factor ,chemistry ,biology.protein ,Signal transduction ,Carrier Proteins ,Proto-Oncogene Proteins c-akt ,Interferon regulatory factors ,Signal Transduction - Abstract
The precise signaling mechanisms by which type II IFN receptors control expression of unique genes to induce biological responses remain to be established. We provide evidence that Sin1, a known element of the mammalian target of rapamycin complex 2 (mTORC2), is required for IFNγ-induced phosphorylation and activation of AKT and that such activation mediates downstream regulation of mTORC1 and its effectors. These events play important roles in the assembly of the eukaryotic translation initiation factor 4F (eIF4F) and mRNA translation of IFN-stimulated genes. Interestingly, IFNγ-induced tyrosine phosphorylation of STAT1 is reduced in cells with targeted disruption of Sin1, leading to decreased transcription of several IFNγ-inducible genes in an mTORC2-independent manner. Additionally, our studies establish that Sin1 is essential for generation of type II IFN-dependent antiviral effects and antiproliferative responses in normal and malignant hematopoiesis. Together, our findings establish an important role for Sin1 in both transcription and translation of IFN-stimulated genes and type II IFN-mediated biological responses, involving both mTORC2-dependent and -independent functions.
- Published
- 2017
23. Interferon-β is a key regulator of proinflammatory events in experimental autoimmune encephalomyelitis
- Author
-
Beata Majchrzak-Kita, Leesa M. Pennell, Reza Rahbar, Eleanor N. Fish, Ehtesham Baig, Thomas T. Murooka, and Carole L. Galligan
- Subjects
Male ,Encephalomyelitis, Autoimmune, Experimental ,Encephalomyelitis ,T cell ,Autoimmunity ,Inflammation ,Cell Separation ,medicine.disease_cause ,Myelin oligodendrocyte glycoprotein ,Mice ,Interferon ,Animals ,Medicine ,Mice, Knockout ,Autoimmune disease ,biology ,business.industry ,Gene Expression Profiling ,Experimental autoimmune encephalomyelitis ,Interferon-beta ,Flow Cytometry ,medicine.disease ,Immunohistochemistry ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Neurology ,Immunology ,biology.protein ,Cytokines ,Th17 Cells ,Female ,Neurology (clinical) ,medicine.symptom ,business ,medicine.drug - Abstract
Background: Interferon (IFN)-β is an effective therapy for relapsing-remitting multiple sclerosis, yet its mechanism of action remains ill-defined. Objectives: Our objective was to characterize the role of IFN-β in immune regulation in experimental autoimmune encephalomyelitis (EAE). Methods: IFN-β +/+ and IFN-β—/— mice were immunized with myelin oligodendrocyte glycoprotein peptide in the presence or absence of IFN-β, to induce EAE. Disease pathogenesis was monitored in the context of incidence, time of onset, clinical score, and immune cell activation in the brains, spleens and lymph nodes of affected mice. Results: Compared with IFN-β+/+ mice, IFN-β—/ — mice exhibited an earlier onset and a more rapid progression of EAE, increased numbers of CD11b+ leukocytes infiltrating affected brains and an increased percentage of Th17 cells in the central nervous system and draining lymph nodes. IFN-β treatment delayed disease onset and reduced disease severity. Ex vivo experiments revealed that the lack of IFN-β results in enhanced generation of autoreactive T cells, a likely consequence of the absence of IFN-β-regulated events in both the CD4+ T cells and antigen-presenting dendritic cells. Gene expression analysis of IFN-β-treated bone marrow macrophages (CD11b +) identified modulation of genes affecting T cell proliferation and Th17 differentiation. Conclusions: We conclude that IFN-β acts to suppress the generation of autoimmune-inducing Th17 cells during the development of disease as well as modulating pro-inflammatory mediators.
- Published
- 2010
- Full Text
- View/download PDF
24. Interferon-Dependent Engagement of Eukaryotic Initiation Factor 4B via S6 Kinase (S6K)- and Ribosomal Protein S6K-Mediated Signals
- Author
-
Leonidas C. Platanias, Sara C. Kozma, Barbara Kroczynska, Eleanor N. Fish, Antonella Sassano, Surinder Kaur, Beata Majchrzak-Kita, and Efstratios Katsoulidis
- Subjects
P70-S6 Kinase 1 ,Biology ,Ribosomal Protein S6 Kinases, 90-kDa ,Interferon-gamma ,Mice ,Eukaryotic initiation factor ,Animals ,Humans ,Eukaryotic Initiation Factors ,RNA, Small Interfering ,EIF4B ,Molecular Biology ,Cells, Cultured ,Mice, Knockout ,EIF4E ,Interferon-alpha ,Ribosomal Protein S6 Kinases, 70-kDa ,Articles ,Cell Biology ,EIF4A1 ,Fibroblasts ,Protein kinase R ,Molecular biology ,Eukaryotic translation initiation factor 4 gamma ,Cell biology ,EIF4EBP1 ,Signal Transduction - Abstract
Although the roles of Jak-Stat pathways in type I and II interferon (IFN)-dependent transcriptional regulation are well established, the precise mechanisms of mRNA translation for IFN-sensitive genes remain to be defined. We examined the effects of IFNs on the phosphorylation/activation of eukaryotic translation initiation factor 4B (eIF4B). Our data show that eIF4B is phosphorylated on Ser422 during treatment of sensitive cells with alpha IFN (IFN-alpha) or IFN-gamma. Such phosphorylation is regulated, in a cell type-specific manner, by either the p70 S6 kinase (S6K) or the p90 ribosomal protein S6K (RSK) and results in enhanced interaction of the protein with eIF3A (p170/eIF3A) and increased associated ATPase activity. Our data also demonstrate that IFN-inducible eIF4B activity and IFN-stimulated gene 15 protein (ISG15) or IFN-gamma-inducible chemokine CXCL-10 protein expression are diminished in S6k1/S6k2 double-knockout mouse embryonic fibroblasts. In addition, IFN-alpha-inducible ISG15 protein expression is blocked by eIF4B or eIF3A knockdown, establishing a requirement for these proteins in mRNA translation/protein expression by IFNs. Importantly, the generation of IFN-dependent growth inhibitory effects on primitive leukemic progenitors is dependent on activation of the S6K/eIF4B or RSK/eIF4B pathway. Taken together, our findings establish critical roles for S6K and RSK in the induction of IFN-dependent biological effects and define a key regulatory role for eIF4B as a common mediator and integrator of IFN-generated signals from these kinases.
- Published
- 2009
- Full Text
- View/download PDF
25. Activation of Protein Kinase Cη by Type I Interferons
- Author
-
Jessica K. Altman, Efstratios Katsoulidis, Antonella Sassano, Amittha Wickrema, Beata Majchrzak-Kita, Eleanor N. Fish, Hui Liu, Amanda J. Redig, and Leonidas C. Platanias
- Subjects
Protein Kinase C-alpha ,Antigens, CD34 ,Protein Kinase C beta ,Biology ,Biochemistry ,Antigens, CD ,Cell Line, Tumor ,Receptors, Transferrin ,medicine ,Animals ,Humans ,RNA, Small Interfering ,Protein kinase A ,Receptor ,Molecular Biology ,Myeloid Progenitor Cells ,Protein Kinase C ,Protein kinase C ,Janus kinase 1 ,Mechanisms of Signal Transduction ,Cell Biology ,Cell biology ,Enzyme Activation ,Isoenzymes ,Leukemia, Myeloid ,Interferon Type I ,Phosphorylation ,Signal transduction ,Interferon type I ,medicine.drug - Abstract
Type I interferons (IFNs) are cytokines with diverse biological properties, including antiviral, growth inhibitory, and immunomodulatory effects. Although several signaling pathways are activated during engagement of the type I IFN receptor and participate in the induction of IFN responses, the mechanisms of generation of specific signals for distinct biological effects remain to be elucidated. We provide evidence that a novel member of the protein kinase C (PKC) family of proteins is rapidly phosphorylated and activated during engagement of the type I IFN receptor. In contrast to other members of the PKC family that are also regulated by IFN receptors, PKCη does not regulate IFN-inducible transcription of interferon-stimulated genes or generation of antiviral responses. However, its function promotes cell cycle arrest and is essential for the generation of the suppressive effects of IFNα on normal and leukemic human myeloid (colony-forming unit-granulocyte macrophage) bone marrow progenitors. Altogether, our studies establish PKCη as a unique element in IFN signaling that plays a key and essential role in the generation of the regulatory effects of type I IFNs on normal and leukemic hematopoiesis.
- Published
- 2009
- Full Text
- View/download PDF
26. De Novo Design of Nonpeptidic Compounds Targeting the Interactions between Interferon-α and its Cognate Cell Surface Receptor
- Author
-
Beata Majchrzak-Kita, Eleanor N. Fish, Lianhu Wei, Angelica M. Bello, Lakshmi P. Kotra, Tanushree Bende, and Xiaoyang Wang
- Subjects
Models, Molecular ,In silico ,Receptor, Interferon alpha-beta ,Thiophenes ,Guanidines ,stat ,Structure-Activity Relationship ,Interferon ,Cell surface receptor ,Cell Line, Tumor ,Drug Discovery ,medicine ,Humans ,Phosphorylation ,Furans ,Receptor ,Chemistry ,Molecular Mimicry ,Interferon-alpha ,DNA ,Small molecule ,Pyrimidines ,STAT1 Transcription Factor ,Biochemistry ,Drug Design ,Molecular Medicine ,Signal transduction ,Peptides ,medicine.drug - Abstract
Type 1 interferons (IFN) bind specifically to the corresponding receptor, IFNAR. Agonists and antagonists for IFNAR have potential therapeutic value in the treatment of viral infections and systemic lupus erythematosus, respectively. Specific sequences on the surface of IFN, IFN receptor recognition peptides (IRRPs) mediate the binding and signal transduction when IFN interacts with IFNAR. Structural features of two such IRRPs, IRRP-1 and IRRP-3, were used as templates to design small molecule mimetics. In silico screening was used to identify the molecular structural features mimicking their surface characteristics. A set of 26 compounds were synthesized and their ability to interfere with IFN-IFNAR interactions was investigated. Two compounds exhibited antagonist activity, specifically, blocking IFN-inducible Stat phosphorylation Stat complex-DNA binding. Design principles revealed here pave the way toward a novel series of small molecules as antagonists for IFN-IFNAR interactions.
- Published
- 2008
- Full Text
- View/download PDF
27. Suppression of Interferon (IFN)-inducible Genes and IFN-mediated Functional Responses in BCR-ABL-expressing Cells
- Author
-
Patrick Yoon, Brian J. Druker, Alison Jordan, Efstratios Katsoulidis, Leonidas C. Platanias, Beata Majchrzak-Kita, Nathalie Carayol, Antonella Sassano, and Eleanor N. Fish
- Subjects
STAT3 Transcription Factor ,Transcription, Genetic ,Response element ,Fusion Proteins, bcr-abl ,Response Elements ,Models, Biological ,Biochemistry ,Mice ,Interferon ,Cell Line, Tumor ,hemic and lymphatic diseases ,medicine ,Animals ,Humans ,STAT1 ,STAT3 ,Molecular Biology ,Cell Proliferation ,biology ,Mechanisms of Signal Transduction ,Promoter ,Cell Biology ,Fusion protein ,STAT1 Transcription Factor ,IRF1 ,Mutation ,Cancer research ,biology.protein ,Tyrosine ,Interferons ,Signal transduction ,Signal Transduction ,medicine.drug - Abstract
The interferons (IFNs) are cytokines that play key roles in host defense against viral infections and immune surveillance against cancer. We report that BCR-ABL transformation of hematopoietic cells results in suppression of IFN-dependent responses, including transcription of IFN-inducible genes and generation of IFN-mediated antiviral effects. BCR-ABL transformation suppresses expression of several IFN-regulated genes containing IFN-sensitive response element (ISRE) or GAS elements in their promoters, including Isg15, Irf1, Irf9, and Ifit2 (interferon-induced protein with tetratricopeptide repeats 2). Suppression of transcription of ISRE-containing genes is also seen in cells expressing various BCR-ABL kinase domain mutants, including T315I, H396P, Y253F, and E255K, but not kinase-defective BCR-ABL. Such effects are associated with impaired IFN-dependent phosphorylation of Stat1 on Tyr701 and Stat3 on Tyr705 and defective binding of Stat complexes to ISRE or GAS elements. Beyond suppression of Stat activities, BCR-ABL inhibits IFN-inducible phosphorylation/activation of the p38 MAPK, suggesting a dual mechanism by which this abnormal fusion protein blocks IFN transcriptional responses. The inhibitory activities of BCR-ABL ultimately result in impaired IFNα-mediated protection against encephalomyocarditis virus infection and reversal of IFN-dependent growth suppression. Altogether, our data provide evidence for a novel mechanism by which BCR-ABL impairs host defenses and promotes malignant transformation, involving dual suppression of IFN-activated signaling pathways.
- Published
- 2008
- Full Text
- View/download PDF
28. A Rapid Screening Assay Identifies Monotherapy with Interferon-ß and Combination Therapies with Nucleoside Analogs as Effective Inhibitors of Ebola Virus
- Author
-
Gary P. Kobinger, Stephen D S McCarthy, Thomas Hoenen, Donald R. Branch, Eleanor N. Fish, Trina Racine, Hannah N Kozlowski, Darren P. Baker, and Beata Majchrzak-Kita
- Subjects
0301 basic medicine ,lcsh:Arctic medicine. Tropical medicine ,lcsh:RC955-962 ,viruses ,030106 microbiology ,Brincidofovir ,Pharmacology ,Favipiravir ,Biology ,medicine.disease_cause ,Virus Replication ,Antiviral Agents ,Virus ,Maraviroc ,03 medical and health sciences ,Zidovudine ,Ebola Hemorrhagic Fever ,Cyclohexanes ,medicine ,Humans ,Ebola virus ,Nucleoside analogue ,lcsh:Public aspects of medicine ,Public Health, Environmental and Occupational Health ,virus diseases ,lcsh:RA1-1270 ,Nucleosides ,Interferon-beta ,Triazoles ,Ebolavirus ,Virology ,3. Good health ,030104 developmental biology ,Infectious Diseases ,Viral replication ,CCR5 Receptor Antagonists ,Toremifene ,medicine.drug ,Research Article - Abstract
To date there are no approved antiviral drugs for the treatment of Ebola virus disease (EVD). While a number of candidate drugs have shown limited efficacy in vitro and/or in non-human primate studies, differences in experimental methodologies make it difficult to compare their therapeutic effectiveness. Using an in vitro model of Ebola Zaire replication with transcription-competent virus like particles (trVLPs), requiring only level 2 biosafety containment, we compared the activities of the type I interferons (IFNs) IFN-α and IFN-ß, a panel of viral polymerase inhibitors (lamivudine (3TC), zidovudine (AZT) tenofovir (TFV), favipiravir (FPV), the active metabolite of brincidofovir, cidofovir (CDF)), and the estrogen receptor modulator, toremifene (TOR), in inhibiting viral replication in dose-response and time course studies. We also tested 28 two- and 56 three-drug combinations against Ebola replication. IFN-α and IFN-ß inhibited viral replication 24 hours post-infection (IC50 0.038μM and 0.016μM, respectively). 3TC, AZT and TFV inhibited Ebola replication when used alone (50–62%) or in combination (87%). They exhibited lower IC50 (0.98–6.2μM) compared with FPV (36.8μM), when administered 24 hours post-infection. Unexpectedly, CDF had a narrow therapeutic window (6.25–25μM). When dosed >50μM, CDF treatment enhanced viral infection. IFN-ß exhibited strong synergy with 3TC (97.3% inhibition) or in triple combination with 3TC and AZT (95.8% inhibition). This study demonstrates that IFNs and viral polymerase inhibitors may have utility in EVD. We identified several 2 and 3 drug combinations with strong anti-Ebola activity, confirmed in studies using fully infectious ZEBOV, providing a rationale for testing combination therapies in animal models of lethal Ebola challenge. These studies open up new possibilities for novel therapeutic options, in particular combination therapies, which could prevent and treat Ebola infection and potentially reduce drug resistance., Author Summary Studies to evaluate the effectiveness of candidate antiviral drugs to inhibit Ebola virus infection have been hampered by the availability and access to level 4 containment facilities. Using a mini-genome model system that generates Ebola virus-like particles that infect cells, we have been able to screen a panel of candidate drugs for antiviral activity, under normal level 2 containment. We compared the activities of 8 different antivirals from 3 drug classes, including drugs repurposed for the treatment of Ebola: type I interferons and nucleoside analogs. Our data indicate that IFN-ß is a potent inhibitor of Ebola virus, contributing to the decision to conduct a clinical trial of IFN-ß treatment for Ebola virus disease in Guinea. Moreover, we identified that 2 and 3 drug combinations inhibit Ebola replication when administered 24 hours post-infection. Drug combinations have important implications for clinical use, since lower doses of each drug are administered, potentially decreasing side-effects and, based on different mechanisms of action, there is less likelihood for the emergence of drug resistance. These studies set the stage for both preclinical and clinical evaluation.
- Published
- 2016
29. CCL5-CCR5-mediated Apoptosis in T Cells
- Author
-
Beata Majchrzak-Kita, Eleanor N. Fish, Mark M. Wong, Amanda E. I. Proudfoot, Thomas T. Murooka, and Ramtin Rahbar
- Subjects
Inhibitor of apoptosis domain ,Glycosaminoglycan binding ,Programmed cell death ,Cytochrome c ,T cell ,virus diseases ,hemic and immune systems ,Cell Biology ,Biology ,Biochemistry ,Molecular biology ,CCL5 ,stomatognathic diseases ,medicine.anatomical_structure ,stomatognathic system ,Cell culture ,Apoptosis ,parasitic diseases ,biology.protein ,medicine ,Molecular Biology - Abstract
CCL5 (RANTES (regulated on activation normal T cell expressed and secreted)) and its cognate receptor, CCR5, have been implicated in T cell activation. CCL5 binding to glycosaminoglycans (GAGs) on the cell surface or in extracellular matrix sequesters CCL5, thereby immobilizing CCL5 to provide the directional signal. In two CCR5-expressing human T cell lines, PM1.CCR5 and MOLT4.CCR5, and in human peripheral blood-derived T cells, micromolar concentrations of CCL5 induce apoptosis. CCL5-induced cell death involves the cytosolic release of cytochrome c, the activation of caspase-9 and caspase-3, and poly(ADP-ribose) polymerase cleavage. CCL5-induced apoptosis is CCR5-dependent, since native PM1 and MOLT4 cells lacking CCR5 expression are resistant to CCL5-induced cell death. Furthermore, we implicate tyrosine 339 as a critical residue involved in CCL5-induced apoptosis, since PM1 cells expressing a tyrosine mutant receptor, CCR5Y339F, do not undergo apoptosis. We show that CCL5-CCR5-mediated apoptosis is dependent on cell surface GAG binding. The addition of exogenous heparin and chondroitin sulfate and GAG digestion from the cell surface protect cells from apoptosis. Moreover, the non-GAG binding variant, (44AANA47)-CCL5, fails to induce apoptosis. To address the role of aggregation in CCL5-mediated apoptosis, nonaggregating CCL5 mutant E66S, which forms dimers, and E26A, which form tetramers at micromolar concentrations, were utilized. Unlike native CCL5, the E66S mutant fails to induce apoptosis, suggesting that tetramers are the minimal higher ordered CCL5 aggregates required for CCL5-induced apoptosis. Viewed altogether, these data suggest that CCL5-GAG binding and CCL5 aggregation are important for CCL5 activity in T cells, specifically in the context of CCR5-mediated apoptosis.
- Published
- 2006
- Full Text
- View/download PDF
30. Critical roles for Rictor/Sin1 complexes in IFN-dependent gene transcription and generation of antiproliferative responses
- Author
-
Eleanor N. Fish, Barbara Kroczynska, Surinder Kaur, Leonidas C. Platanias, Antonella Sassano, Raffaele A. Calogero, Bing Su, Brady L. Stein, Brandon McMahon, Jessica K. Altman, Beata Majchrzak-Kita, Bhumika Sharma, and Ahmet Dirim Arslan
- Subjects
Cell signaling ,Transcription, Genetic ,Antineoplastic Agents ,Biology ,RICTOR Gene ,Biochemistry ,stat ,Mice ,medicine ,Animals ,Humans ,Phosphorylation ,Molecular Biology ,Polycythemia Vera ,Cells, Cultured ,Regulation of gene expression ,Gene knockdown ,digestive, oral, and skin physiology ,JAK-STAT signaling pathway ,Cell Biology ,Fibroblasts ,Hematopoietic Stem Cells ,Molecular biology ,Cell biology ,Gene Expression Regulation, Neoplastic ,Rapamycin-Insensitive Companion of mTOR Protein ,Gene Knockdown Techniques ,Interferon Type I ,Signal transduction ,Carrier Proteins ,Interferon type I ,medicine.drug ,Signal Transduction - Abstract
We provide evidence that type I IFN-induced STAT activation is diminished in cells with targeted disruption of the Rictor gene, whose protein product is a key element of mTOR complex 2. Our studies show that transient or stable knockdown of Rictor or Sin1 results in defects in activation of elements of the STAT pathway and reduced STAT-DNA binding complexes. This leads to decreased expression of several IFN-inducible genes that mediate important biological functions. Our studies also demonstrate that Rictor and Sin1 play essential roles in the generation of the suppressive effects of IFNα on malignant erythroid precursors from patients with myeloproliferative neoplasms. Altogether, these findings provide evidence for critical functions for Rictor/Sin1 complexes in type I IFN signaling and the generation of type I IFN antineoplastic responses.
- Published
- 2014
31. Regulatory effects of mTORC2 complexes in type I IFN signaling and in the generation of IFN responses
- Author
-
Eleanor N. Fish, Bing Su, Antonella Sassano, Surinder Kaur, Darren P. Baker, Beata Majchrzak-Kita, and Leonidas C. Platanias
- Subjects
Immunoblotting ,mTORC1 ,Mechanistic Target of Rapamycin Complex 2 ,Biology ,mTORC2 ,Mice ,Animals ,Humans ,Phosphorylation ,Luciferases ,Protein kinase B ,Regulation of gene expression ,Ribosomal Protein S6 ,Multidisciplinary ,Effector ,Reverse Transcriptase Polymerase Chain Reaction ,Ribosomal Protein S6 Kinases ,TOR Serine-Threonine Kinases ,Biological Sciences ,Cell biology ,Rapamycin-Insensitive Companion of mTOR Protein ,Gene Expression Regulation ,Ribosomal protein s6 ,Multiprotein Complexes ,Trans-Activators ,Interferons ,Signal transduction ,Carrier Proteins ,Proto-Oncogene Proteins c-akt ,HeLa Cells ,Signal Transduction - Abstract
IFNs transduce signals by binding to cell surface receptors and activating cellular pathways and regulatory networks that control transcription of IFN-stimulated genes (ISGs) and mRNA translation, leading to generation of protein products that mediate biological responses. Previous studies have shown that type I IFN receptor-engaged pathways downstream of AKT and mammalian target of rapamycin complex (mTORC) 1 play important roles in mRNA translation of ISGs and the generation of IFN responses, but the roles of mTORC2 complexes in IFN signaling are unknown. We provide evidence that mTORC2 complexes control IFN-induced phosphorylation of AKT on serine 473 and their function is ultimately required for IFN-dependent gene transcription via interferon-stimulated response elements. We also demonstrate that such complexes exhibit regulatory effects on other IFN-dependent mammalian target of rapamycin-mediated signaling events, likely via engagement of the AKT/mTORC1 axis, including IFN-induced phosphorylation of S6 kinase and its effector rpS6, as well as phosphorylation of the translational repressor 4E-binding protein 1. We also show that induction of ISG protein expression and the generation of antiviral responses are defective in Rictor and mLST8-KO cells. Together, our data provide evidence for unique functions of mTORC2 complexes in the induction of type I IFN responses and suggest a critical role for mTORC2-mediated signals in IFN signaling.
- Published
- 2012
32. Dynamic accumulation of plasmacytoid dendritic cells in lymph nodes is regulated by interferon-beta
- Author
-
Eleanor N. Fish, Yunfei Gao, Jennifer L. Gommerman, and Beata Majchrzak-Kita
- Subjects
Antigens, Differentiation, T-Lymphocyte ,Lymphoid Tissue ,medicine.medical_treatment ,Immunology ,Plasmacytoid dendritic cell ,Biology ,Biochemistry ,Mice ,Antigen ,Orthomyxoviridae Infections ,Interferon ,Antigens, CD ,Cell Movement ,medicine ,Animals ,Lectins, C-Type ,Antigen-presenting cell ,Cells, Cultured ,Mice, Knockout ,Interferon-alpha ,hemic and immune systems ,Cell Biology ,Hematology ,Dendritic cell ,Dendritic Cells ,Interferon-beta ,Orthomyxoviridae ,Recombinant Proteins ,Cell biology ,Immunosurveillance ,Mice, Inbred C57BL ,Cytokine ,Lymph Nodes ,Ex vivo ,medicine.drug - Abstract
Plasmacytoid dendritic cells (pDCs) represent a major cellular component of our front-line defense against viruses because of their capacity to rapidly secrete type I interferon (IFN)–α and -β after infection. Constant immunosurveillance of the host requires that lymphocytes traffic through lymph nodes (LNs) to sample antigen, yet little is known about the dynamics of pDC accumulation within the secondary lymphoid organs. Here we show that pDCs readily accumulate within the secondary lymphoid organs of mice after virus infection. Interestingly, retention of pDC within LNs is enhanced in the presence of the sphingoshine-1-phosphate receptor agonist FTY720 in a manner similar to that observed for B and T lymphocytes. Ex vivo comparison of mouse pDCs with lymphocytes revealed that pDCs express sphingoshine-1-phosphate 4 and also constitutively express CD69, which is further up-regulated upon virus infection. In IFN-β−/− mice, accumulation of pDC and lymphocytes within LNs is reduced both during viral infection and under steady state conditions, and these defects can be reversed by adding recombinant IFN-β in vivo. These data suggest that pDC and lymphocytes use similar mechanisms for retention within LNs and that these processes are influenced by IFN-β even in the absence of viral infection.
- Published
- 2009
33. Role of Schlafen 2 (SLFN2) in the Generation of Interferon α-induced Growth Inhibitory Responses*
- Author
-
Nathalie Carayol, Eleanor N. Fish, Beata Majchrzak-Kita, Leonidas C. Platanias, Alison Jordan, Iwona M. Konieczna, Jennifer Woodard, Elizabeth A. Eklund, Efstratios Katsoulidis, and Antonella Sassano
- Subjects
MAPK/ERK pathway ,MAP Kinase Signaling System ,Cell Cycle Proteins ,Mice, Transgenic ,Biology ,Biochemistry ,p38 Mitogen-Activated Protein Kinases ,stat ,Cell Line ,Mice ,Animals ,Progenitor cell ,Molecular Biology ,Cell Proliferation ,Gene knockdown ,Cell growth ,Mechanisms of Signal Transduction ,Cell Cycle ,Interferon-alpha ,Cell Biology ,Cell cycle ,Hematopoietic Stem Cells ,Cell biology ,STAT1 Transcription Factor ,Cell culture ,STAT protein ,NIH 3T3 Cells ,Cytokines ,CpG Islands - Abstract
The precise STAT-regulated gene targets that inhibit cell growth and generate the antitumor effects of Type I interferons (IFNs) remain unknown. We provide evidence that Type I IFNs regulate expression of Schlafens (SLFNs), a group of genes involved in the control of cell cycle progression and growth inhibitory responses. Using cells with targeted disruption of different STAT proteins and/or the p38 MAP kinase, we demonstrate that the IFN-dependent expression of distinct Schlafen genes is differentially regulated by STAT complexes and the p38 MAP kinase pathway. We also provide evidence for a key functional role of a member of the SLFN family, SLFN2, in the induction of the growth-suppressive effects of IFNs. This is shown in studies demonstrating that knockdown of SLFN2 enhances hematopoietic progenitor colony formation and reverses the growth-suppressive effects of IFNalpha on normal hematopoietic progenitors. Importantly, NIH3T3 or L929 cells with stable knockdown of SLFN2 form more colonies in soft agar, implicating this protein in the regulation of anchorage-independent growth. Altogether, our data implicate SLFN2 as a negative regulator of the metastatic and growth potential of malignant cells and strongly suggest a role for the SLFN family of proteins in the generation of the antiproliferative effects of Type I IFNs.
- Published
- 2009
34. Regulatory effects of mammalian target of rapamycin-activated pathways in type I and II interferon signaling
- Author
-
Eleanor N. Fish, Emmanuel Petroulakis, Darren P. Baker, Beata Majchrzak-Kita, Lakhvir Lal, Leonidas C. Platanias, Nahum Sonenberg, Surinder Kaur, Maya Srikanth, Antonella Sassano, and Nissim Hay
- Subjects
Cell Cycle Proteins ,Biology ,Biochemistry ,Antiviral Agents ,Tuberous Sclerosis Complex 1 Protein ,Interferon-gamma ,Mice ,Tuberous Sclerosis Complex 2 Protein ,Transcriptional regulation ,Animals ,Humans ,Molecular Biology ,PI3K/AKT/mTOR pathway ,Adaptor Proteins, Signal Transducing ,Regulation of gene expression ,TOR Serine-Threonine Kinases ,Tumor Suppressor Proteins ,Signal transducing adaptor protein ,Cell Biology ,EIF4A1 ,Phosphoproteins ,Molecular biology ,Cell biology ,Chemokine CXCL10 ,Gene Expression Regulation ,Interferon Type I ,Phosphorylation ,TSC2 ,Signal transduction ,Chemokines, CXC ,Protein Kinases ,Signal Transduction - Abstract
The mechanisms regulating initiation of mRNA translation for the generation of protein products that mediate interferon (IFN) responses are largely unknown. We have previously shown that both Type I and II IFNs engage the mammalian target of rapamycin (mTOR), resulting in downstream phosphorylation and deactivation of the translational repressor 4E-BP1 (eIF4E-binding protein 1). In the current study, we provide direct evidence that such regulation of 4E-BP1 by IFNalpha or IFNgamma results in sequential dissociation of 4E-BP1 from eukaryotic initiation factor-4E and subsequent formation of a functional complex between eukaryotic initiation factor-4E and eukaryotic initiation factor-4G, to allow initiation of mRNA translation. We also demonstrate that the induction of key IFNalpha- or IFNgamma-inducible proteins (ISG15 (interferon-stimulated gene 15) and CXCL10) that mediate IFN responses are enhanced in 4E-BP1 (4E-BP1(-/-)) knockout MEFs, as compared with wild-type 4E-BP1(+/+) MEFs. On the other hand, IFN-dependent transcriptional regulation of the Isg15 and Cxcl10 genes is intact in the absence of 4E-BP1, as determined by real time reverse transcriptase-PCR assays and promoter assays for ISRE and GAS, establishing that 4E-BP1 plays a selective negative regulatory role in IFN-induced mRNA translation. Interestingly, the induction of expression of ISG15 and CXCL10 proteins by IFNs was also strongly enhanced in cells lacking expression of the tuberin (TSC2(-/-)) or hamartin (TSC1(-/-)) genes, consistent with the known negative regulatory effect of the TSC1-TSC2 complex on mTOR activation. In other work, we demonstrate that the induction of an IFN-dependent antiviral response is strongly enhanced in cells lacking expression of 4E-BP1 and TSC2, demonstrating that these elements of the IFN-activated mTOR pathway exhibit important regulatory effects in the generation of IFN responses. Taken altogether, our data suggest an important role for mTOR-dependent pathways in IFN signaling and identify 4E-BP1 and TSC1-TSC2 as key components in the generation of IFN-dependent biological responses.
- Published
- 2006
35. CCL5-CCR5-mediated apoptosis in T cells: Requirement for glycosaminoglycan binding and CCL5 aggregation
- Author
-
Thomas T, Murooka, Mark M, Wong, Ramtin, Rahbar, Beata, Majchrzak-Kita, Amanda E I, Proudfoot, and Eleanor N, Fish
- Subjects
Receptors, CCR5 ,Caspase 3 ,T-Lymphocytes ,Cell Membrane ,Cytochromes c ,Apoptosis ,Caspase 9 ,Extracellular Matrix ,Caspases ,Cell Line, Tumor ,Chemokines, CC ,Humans ,Poly(ADP-ribose) Polymerases ,Chemokine CCL5 ,Glycosaminoglycans ,Protein Binding - Abstract
CCL5 (RANTES (regulated on activation normal T cell expressed and secreted)) and its cognate receptor, CCR5, have been implicated in T cell activation. CCL5 binding to glycosaminoglycans (GAGs) on the cell surface or in extracellular matrix sequesters CCL5, thereby immobilizing CCL5 to provide the directional signal. In two CCR5-expressing human T cell lines, PM1.CCR5 and MOLT4.CCR5, and in human peripheral blood-derived T cells, micromolar concentrations of CCL5 induce apoptosis. CCL5-induced cell death involves the cytosolic release of cytochrome c, the activation of caspase-9 and caspase-3, and poly(ADP-ribose) polymerase cleavage. CCL5-induced apoptosis is CCR5-dependent, since native PM1 and MOLT4 cells lacking CCR5 expression are resistant to CCL5-induced cell death. Furthermore, we implicate tyrosine 339 as a critical residue involved in CCL5-induced apoptosis, since PM1 cells expressing a tyrosine mutant receptor, CCR5Y339F, do not undergo apoptosis. We show that CCL5-CCR5-mediated apoptosis is dependent on cell surface GAG binding. The addition of exogenous heparin and chondroitin sulfate and GAG digestion from the cell surface protect cells from apoptosis. Moreover, the non-GAG binding variant, (44AANA47)-CCL5, fails to induce apoptosis. To address the role of aggregation in CCL5-mediated apoptosis, nonaggregating CCL5 mutant E66S, which forms dimers, and E26A, which form tetramers at micromolar concentrations, were utilized. Unlike native CCL5, the E66S mutant fails to induce apoptosis, suggesting that tetramers are the minimal higher ordered CCL5 aggregates required for CCL5-induced apoptosis. Viewed altogether, these data suggest that CCL5-GAG binding and CCL5 aggregation are important for CCL5 activity in T cells, specifically in the context of CCR5-mediated apoptosis.
- Published
- 2006
36. 1 Dual regulatory roles of the phosphatidylinositol 3-kinase in interferon signaling
- Author
-
Saskia M. Brachmann, Ajith M. Joseph, Beata Majchrzak-Kita, Elizabeth A. Eklund, Antonella Sassano, Amit Verma, Leonidas C. Platanias, Eleanor N. Fish, and Surinder Kaur
- Subjects
Kinase ,Chemistry ,Akt/PKB signaling pathway ,Immunology ,Hematology ,DUAL (cognitive architecture) ,Biochemistry ,Cell biology ,Pleckstrin homology domain ,chemistry.chemical_compound ,Interferon ,medicine ,Immunology and Allergy ,Phosphatidylinositol ,Molecular Biology ,Phosphoinositide-dependent kinase-1 ,Interferon regulatory factors ,medicine.drug - Published
- 2008
- Full Text
- View/download PDF
37. De Novo Design of Nonpeptidic Compounds Targeting the Interactions between Interferon-α and its Cognate Cell Surface Receptor.
- Author
-
Angelica M. Bello, Tanushree Bende, Lianhu Wei, Xiaoyang Wang, Beata Majchrzak-Kita, Eleanor N. Fish, and Lakshmi P. Kotra
- Published
- 2008
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.