30 results on '"Kathryn L. Pellegrini"'
Search Results
2. Modulation of type I interferon responses potently inhibits SARS-CoV-2 replication and inflammation in rhesus macaques
- Author
-
Timothy N. Hoang, Elise G. Viox, Amit A. Upadhyay, Zachary Strongin, Gregory K. Tharp, Maria Pino, Rayhane Nchioua, Maximilian Hirschenberger, Matthew Gagne, Kevin Nguyen, Justin L. Harper, Shir Marciano, Arun K. Boddapati, Kathryn L. Pellegrini, Jennifer Tisoncik-Go, Leanne S. Whitmore, Kirti A. Karunakaran, Melissa Roy, Shannon Kirejczyk, Elizabeth H. Curran, Chelsea Wallace, Jennifer S. Wood, Fawn Connor-Stroud, Sudhir P. Kasturi, Rebecca D. Levit, Michael Gale, Thomas H. Vanderford, Guido Silvestri, Kathleen Busman-Sahay, Jacob D. Estes, Monica Vaccari, Daniel C. Douek, Konstantin M.J. Sparrer, Frank Kirchhoff, R. Paul Johnson, Gideon Schreiber, Steven E. Bosinger, and Mirko Paiardini
- Abstract
Type-I interferons (IFN-I) are critical mediators of innate control of viral infections, but also drive recruitment of inflammatory cells to sites of infection, a key feature of severe COVID-19. Here, and for the first time, IFN-I signaling was modulated in rhesus macaques (RMs) prior to and during acute SARS-CoV-2 infection using a mutated IFNα2 (IFN-modulator; IFNmod), which has previously been shown to reduce the binding and signaling of endogenous IFN-I. In SARS-CoV-2-infected RMs, IFNmod reduced both antiviral and inflammatory ISGs. Notably, IFNmod treatment resulted in a potent reduction in (i) SARS-CoV-2 viral load in Bronchoalveolar lavage (BAL), upper airways, lung, and hilar lymph nodes; (ii) inflammatory cytokines, chemokines, and CD163+MRC1-inflammatory macrophages in BAL; and (iii) expression of Siglec-1, which enhances SARS-CoV-2 infection and predicts disease severity, on circulating monocytes. In the lung, IFNmod also reduced pathogenesis and attenuated pathways of inflammasome activation and stress response during acute SARS-CoV-2 infection. This study, using an intervention targeting both IFN-α and IFN-β pathways, shows that excessive inflammation driven by type 1 IFN critically contributes to SARS-CoV-2 pathogenesis in RMs, and demonstrates the potential of IFNmod to limit viral replication, SARS-CoV-2 induced inflammation, and COVID-19 severity.
- Published
- 2022
- Full Text
- View/download PDF
3. Systems vaccinology of the BNT162b2 mRNA vaccine in humans
- Author
-
Meera Trisal, Prabhu S. Arunachalam, Bali Pulendran, Thomas Hagan, Allan Feng, Lilit Grigoryan, Natalia Sigal, Sheena Gupta, Kathryn L. Pellegrini, Mark Roskey, Kari C. Nadeau, Sarah Esther Chang, Steven E. Bosinger, Mehul S. Suthar, Sharon Chinthrajah, Venkata Viswanadh Edara, Mihir Shah, Mark M. Davis, Vamsee Mallajosyula, Fei Gao, Sayantani B. Sindher, Florian Wimmers, Yupeng Feng, Scott D. Boyd, Alexandra S. Lee, Holden T. Maecker, Sydney Hamilton, Chunfeng Li, Madeleine K D Scott, Lilin Lai, Purvesh Khatri, Sofia Maysel-Auslender, Shaurya Dhingra, Hadj Aoued, Paul J. Utz, Sangeeta Kowli, Gregory K. Tharp, and Kevin Hrusovsky
- Subjects
Vaccination ,Multidisciplinary ,Innate immune system ,Immune system ,Immunization ,biology ,Immunity ,Immunology ,biology.protein ,biochemical phenomena, metabolism, and nutrition ,Antibody ,Neutralizing antibody ,Acquired immune system - Abstract
The emergency use authorization of two mRNA vaccines in less than a year from the emergence of SARS-CoV-2 represents a landmark in vaccinology1,2. Yet, how mRNA vaccines stimulate the immune system to elicit protective immune responses is unknown. Here we used a systems vaccinology approach to comprehensively profile the innate and adaptive immune responses of 56 healthy volunteers who were vaccinated with the Pfizer–BioNTech mRNA vaccine (BNT162b2). Vaccination resulted in the robust production of neutralizing antibodies against the wild-type SARS-CoV-2 (derived from 2019-nCOV/USA_WA1/2020) and, to a lesser extent, the B.1.351 strain, as well as significant increases in antigen-specific polyfunctional CD4 and CD8 T cells after the second dose. Booster vaccination stimulated a notably enhanced innate immune response as compared to primary vaccination, evidenced by (1) a greater frequency of CD14+CD16+ inflammatory monocytes; (2) a higher concentration of plasma IFNγ; and (3) a transcriptional signature of innate antiviral immunity. Consistent with these observations, our single-cell transcriptomics analysis demonstrated an approximately 100-fold increase in the frequency of a myeloid cell cluster enriched in interferon-response transcription factors and reduced in AP-1 transcription factors, after secondary immunization. Finally, we identified distinct innate pathways associated with CD8 T cell and neutralizing antibody responses, and show that a monocyte-related signature correlates with the neutralizing antibody response against the B.1.351 variant. Collectively, these data provide insights into the immune responses induced by mRNA vaccination and demonstrate its capacity to prime the innate immune system to mount a more potent response after booster immunization. Profiling the immune responses of 56 volunteers vaccinated with BNT162b2 reveals how this mRNA vaccine primes the innate immune system to mount a potent response to SARS-CoV-2 after booster immunization.
- Published
- 2021
- Full Text
- View/download PDF
4. CCR2 Signaling Restricts SARS-CoV-2 Infection
- Author
-
Steven E. Bosinger, Fengzhi Jin, Pei Yong Shi, Jacob E. Kohlmeier, Jeronay Thomas, Vineet D. Menachery, Abigail Vanderheiden, Robyn S. Klein, David A Cowan, Kathryn L. Pellegrini, Meredith E. Davis-Gardner, Arash Grakoui, Allison Soung, Katharine Floyd, and Mehul S. Suthar
- Subjects
Receptors, CCR2 ,medicine.medical_treatment ,viruses ,mouse model ,Pneumonia, Viral ,Inflammation ,Biology ,Virus Replication ,Microbiology ,Proinflammatory cytokine ,Mice ,Virology ,medicine ,Animals ,Lung ,innate immunity ,Innate immune system ,SARS-CoV-2 ,Respiratory disease ,lung inflammation ,COVID-19 ,Viral Load ,respiratory system ,medicine.disease ,Immunity, Innate ,QR1-502 ,respiratory tract diseases ,Mice, Inbred C57BL ,Disease Models, Animal ,Cytokine ,medicine.anatomical_structure ,Immunology ,Cytokines ,Female ,medicine.symptom ,monocytes ,Viral load ,Respiratory tract ,Signal Transduction ,Research Article - Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused a historic pandemic of respiratory disease (coronavirus disease 2019 [COVID-19]), and current evidence suggests that severe disease is associated with dysregulated immunity within the respiratory tract. However, the innate immune mechanisms that mediate protection during COVID-19 are not well defined. Here, we characterize a mouse model of SARS-CoV-2 infection and find that early CCR2 signaling restricts the viral burden in the lung. We find that a recently developed mouse-adapted SARS-CoV-2 (MA-SARS-CoV-2) strain as well as the emerging B.1.351 variant trigger an inflammatory response in the lung characterized by the expression of proinflammatory cytokines and interferon-stimulated genes. Using intravital antibody labeling, we demonstrate that MA-SARS-CoV-2 infection leads to increases in circulating monocytes and an influx of CD45+ cells into the lung parenchyma that is dominated by monocyte-derived cells. Single-cell RNA sequencing (scRNA-Seq) analysis of lung homogenates identified a hyperinflammatory monocyte profile. We utilize this model to demonstrate that mechanistically, CCR2 signaling promotes the infiltration of classical monocytes into the lung and the expansion of monocyte-derived cells. Parenchymal monocyte-derived cells appear to play a protective role against MA-SARS-CoV-2, as mice lacking CCR2 showed higher viral loads in the lungs, increased lung viral dissemination, and elevated inflammatory cytokine responses. These studies have identified a potential CCR2-monocyte axis that is critical for promoting viral control and restricting inflammation within the respiratory tract during SARS-CoV-2 infection. IMPORTANCE SARS-CoV-2 has caused a historic pandemic of respiratory disease (COVID-19), and current evidence suggests that severe disease is associated with dysregulated immunity within the respiratory tract. However, the innate immune mechanisms that mediate protection during COVID-19 are not well defined. Here, we characterize a mouse model of SARS-CoV-2 infection and find that early CCR2-dependent infiltration of monocytes restricts the viral burden in the lung. We find that SARS-CoV-2 triggers an inflammatory response in the lung characterized by the expression of proinflammatory cytokines and interferon-stimulated genes. Using RNA sequencing and flow cytometry approaches, we demonstrate that SARS-CoV-2 infection leads to increases in circulating monocytes and an influx of CD45+ cells into the lung parenchyma that is dominated by monocyte-derived cells. Mechanistically, CCR2 signaling promoted the infiltration of classical monocytes into the lung and the expansion of monocyte-derived cells. Parenchymal monocyte-derived cells appear to play a protective role against MA-SARS-CoV-2, as mice lacking CCR2 showed higher viral loads in the lungs, increased lung viral dissemination, and elevated inflammatory cytokine responses. These studies have identified that the CCR2 pathway is critical for promoting viral control and restricting inflammation within the respiratory tract during SARS-CoV-2 infection.
- Published
- 2021
- Full Text
- View/download PDF
5. TREM2+ and interstitial macrophages orchestrate airway inflammation in SARS-CoV-2 infection in rhesus macaques
- Author
-
Zachary Strongin, Anne Piantadosi, Kevin Nguyen, Susan Pereira Ribeiro, Sydney Hamilton, Thomas H. Vanderford, Rebecca D. Levit, David A. Cowan, Elise G. Viox, Amit A. Upadhyay, Steven E. Bosinger, Jacqueline Corry, Justin L. Harper, Timothy N. Hoang, Rama Rao Amara, Michelle Y.H. Lee, Kathryn L. Pellegrini, Rafick Pierre Sekaly, Tristan R. Horton, Maria Pino, Mirko Paiardini, Arun K. Boddapati, Hadj Aoued, Simon M. Barratt-Boyes, Raymond F. Schinazi, Gregory K. Tharp, and Elizabeth N. Beagle
- Subjects
education.field_of_study ,Innate immune system ,business.industry ,medicine.medical_treatment ,Population ,Inflammation ,respiratory system ,Article ,Proinflammatory cytokine ,Interleukin 10 ,Cytokine ,Immunology ,Medicine ,Macrophage ,medicine.symptom ,business ,education ,Natural killer cell activation - Abstract
The COVID-19 pandemic remains a global health crisis, yet, the immunopathological mechanisms driving the development of severe disease remain poorly defined. Here, we utilize a rhesus macaque (RM) model of SARS-CoV-2 infection to delineate perturbations in the innate immune system during acute infection using an integrated systems analysis. We found that SARS-CoV-2 initiated a rapid infiltration (two days post infection) of plasmacytoid dendritic cells into the lower airway, commensurate with IFNA production, natural killer cell activation, and induction of interferon-stimulated genes. At this early interval, we also observed a significant increase of blood CD14-CD16+ monocytes. To dissect the contribution of lung myeloid subsets to airway inflammation, we generated a novel compendium of RM-specific lung macrophage gene expression using a combination of sc-RNA-Seq data and bulk RNA-Seq of purified populations under steady state conditions. Using these tools, we generated a longitudinal sc-RNA-seq dataset of airway cells in SARS-CoV-2-infected RMs. We identified that SARS-CoV-2 infection elicited a rapid recruitment of two subsets of macrophages into the airway: a C206+MRC1-population resembling murine interstitial macrophages, and a TREM2+ population consistent with CCR2+ infiltrating monocytes, into the alveolar space. These subsets were the predominant source of inflammatory cytokines, accounting for ~75% of IL6 and TNF production, and >90% of IL10 production, whereas the contribution of CD206+MRC+ alveolar macrophages was significantly lower. Treatment of SARS-CoV-2 infected RMs with baricitinib (Olumiant®), a novel JAK1/2 inhibitor that recently received Emergency Use Authorization for the treatment of hospitalized COVID-19 patients, was remarkably effective in eliminating the influx of infiltrating, non-alveolar macrophages in the alveolar space, with a concomitant reduction of inflammatory cytokines. This study has delineated the major subsets of lung macrophages driving inflammatory and anti-inflammatory cytokine production within the alveolar space during SARS-CoV-2 infection.One sentence summaryMulti-omic analyses of hyperacute SARS-CoV-2 infection in rhesus macaques identified two population of infiltrating macrophages, as the primary orchestrators of inflammation in the lower airway that can be successfully treated with baricitinib
- Published
- 2021
- Full Text
- View/download PDF
6. CCR2-dependent monocyte-derived cells restrict SARS-CoV-2 infection
- Author
-
Arash Grakoui, Adrian Creanga, Alexandrine Derrien-Colemyn, Cowan Da, Pei Yong Shi, Meredith E. Davis-Gardner, Vineet D. Menachery, Abigail Vanderheiden, Robyn S. Klein, Jacob E. Kohlmeier, Steven E. Bosinger, Kathryn L. Pellegrini, Amarendra Pegu, Mehul S. Suthar, James W. Thomas, Katharine Floyd, Jin F, and Allison Soung
- Subjects
Lung ,Innate immune system ,Monocyte ,medicine.medical_treatment ,Respiratory disease ,Inflammation ,Biology ,respiratory system ,medicine.disease ,Article ,respiratory tract diseases ,medicine.anatomical_structure ,Cytokine ,Immunology ,medicine ,medicine.symptom ,Viral load ,Respiratory tract - Abstract
SARS-CoV-2 has caused a historic pandemic of respiratory disease (COVID-19) and current evidence suggests severe disease is associated with dysregulated immunity within the respiratory tract. However, the innate immune mechanisms that mediate protection during COVID-19 are not well defined. Here we characterize a mouse model of SARS-CoV-2 infection and find that early CCR2-dependent infiltration of monocytes restricts viral burden in the lung. We find that a recently developed mouse-adapted MA-SARS-CoV-2 strain, as well as the emerging B. 1.351 variant, trigger an inflammatory response in the lung characterized by expression of pro-inflammatory cytokines and interferon-stimulated genes. scRNA-seq analysis of lung homogenates identified a hyper-inflammatory monocyte profile. Using intravital antibody labeling, we demonstrate that MA-SARS-CoV-2 infection leads to increases in circulating monocytes and an influx of CD45+ cells into the lung parenchyma that is dominated by monocyte-derived cells. We utilize this model to demonstrate that mechanistically, CCR2 signaling promotes infiltration of classical monocytes into the lung and expansion of monocyte-derived cells. Parenchymal monocyte-derived cells appear to play a protective role against MA-SARS-CoV-2, as mice lacking CCR2 showed higher viral loads in the lungs, increased lung viral dissemination, and elevated inflammatory cytokine responses. These studies have identified a CCR2-monocyte axis that is critical for promoting viral control and restricting inflammation within the respiratory tract during SARS-CoV-2 infection.
- Published
- 2021
7. A modified vaccinia Ankara vector-based vaccine protects macaques from SARS-CoV-2 infection, immune pathology, and dysfunction in the lungs
- Author
-
Shelly Wang, Steven E. Bosinger, Joyce Cohen, Sally Shin, Pei Yong Shi, Stephanie Fischinger, Fawn Connor-Stroud, Amit A. Upadhyay, Sailaja Gangadhara, Katharine Floyd, Sanjeev Gumber, Narayanaiah Cheedarla, Jennifer S. Wood, Mehul S. Suthar, Rama Rao Amara, Anusmita Sahoo, Ayalnesh Shiferaw, Nanda Kishore Routhu, Rachelle L. Stammen, Venkata Viswanadh Edara, David C. Montefiori, Vineet D. Menachery, Venkata S. Bollimpelli, Shannon Kirejczyk, Caroline Atyeo, Galit Alter, Thomas H. Vanderford, Lilin Lai, Sherrie Jean, Sheikh Abdul Rahman, Kathryn L. Pellegrini, and Arun K. Boddapati
- Subjects
0301 basic medicine ,Modified vaccinia Ankara ,COVID-19 Vaccines ,viruses ,Genetic Vectors ,Immunology ,Gene Expression ,Vaccinia virus ,Antibodies, Viral ,complex mixtures ,Article ,Immunophenotyping ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Immune system ,T-Lymphocyte Subsets ,Gene Order ,Macrophages, Alveolar ,medicine ,Vaccines, DNA ,Animals ,Immunology and Allergy ,Vector (molecular biology) ,Neutralizing antibody ,Antigens, Viral ,Lung ,B cell ,biology ,SARS-CoV-2 ,Vaccination ,COVID-19 ,Virology ,Antibodies, Neutralizing ,respiratory tract diseases ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Infectious Diseases ,Viral replication ,030220 oncology & carcinogenesis ,Spike Glycoprotein, Coronavirus ,biology.protein ,Macaca ,Antibody - Abstract
A combination of vaccination approaches will likely be necessary to fully control the SARS-CoV-2 pandemic. Here, we show that modified vaccinia Ankara (MVA) vectors expressing membrane anchored pre-fusion stabilized spike (MVA/S), but not secreted S1, induced strong neutralizing antibody responses against SARS-CoV-2 in mice. In macaques, the MVA/S vaccination induced strong neutralizing antibodies and CD8+ T cell responses, and showed protection from SARS-CoV-2 infection and virus replication in the lung as early as day 2 following intranasal or intratracheal challenge. Single-cell RNA sequencing analysis of lung cells at day 4 post-infection revealed that MVA/S vaccination also protected macaques from infection-induced inflammation and B cell abnormalities, and lowered induction of interferon stimulated genes. These results demonstrate that MVA/S vaccination induces both neutralizing antibodies and CD8+ T cells in the blood and lung and serves as a potential vaccine candidate against SARS-CoV-2., Graphical Abstract, Modified vaccinia Ankara (MVA) vector-based vaccines are attractive for their excellent safety and ability to induce long-lived humoral and cellular immunity in humans. Routhu et al. show that an MVA-based COVID-19 vaccine encoding prefusion-stabilized spike MVA/S induces strong neutralizing antibody and CD8+ T cell responses and protects macaques from SARS-CoV2 infection, immunopathology and infection-induced B cell abnormalities in the lung.
- Published
- 2021
- Full Text
- View/download PDF
8. Baricitinib treatment resolves lower airway inflammation and neutrophil recruitment in SARS-CoV-2-infected rhesus macaques
- Author
-
Amit A. Upadhyay, Sudhir Pai Kasturi, Elise G. Viox, Steven E. Bosinger, Ernestine A. Mahar, Michael N. Sayegh, Sanjeev Gumber, Justin L. Harper, Arun K. Boddapati, Sijia Tao, Shelly Wang, Rebecca D. Levit, Thomas H. Vanderford, Michelle Y.H. Lee, Shannon Kirejczyk, Sherrie Jean, Keivan Zandi, Susan Pereira Ribeiro, Carly E. Starke, Lanfang Wang, Daniela Weiskopf, Maria Pino, Joyce Cohen, Timothy N. Hoang, Tristan R. Horton, Gregory K. Tharp, Jesse J. Waggoner, Kathleen Busman-Sahay, Anne Piantadosi, Olivia M. Delmas, Kathryn L. Pellegrini, Jennifer S. Wood, Jacob D. Estes, Peter D. Filev, Mirko Paiardini, Rachelle L. Stammen, Elizabeth N. Beagle, Zachary Strongin, Raymond F. Schinazi, Rafick Pierre Sekaly, Kimberly A. Cooney, Fawn Connor-Stroud, and Hilmi Al Shakhshir
- Subjects
Chemokine ,Lung ,biology ,business.industry ,T cell ,Inflammation ,biology.organism_classification ,Systemic inflammation ,Article ,Rhesus macaque ,medicine.anatomical_structure ,Immunology ,medicine ,biology.protein ,Alveolar macrophage ,medicine.symptom ,Viral shedding ,business - Abstract
Effective therapeutics aimed at mitigating COVID-19 symptoms are urgently needed. SARS-CoV-2 induced hypercytokinemia and systemic inflammation are associated with disease severity. Baricitinib, a clinically approved JAK1/2 inhibitor with potent anti-inflammatory properties is currently being investigated in COVID-19 human clinical trials. Recent reports suggest that baricitinib may also have antiviral activity in limiting viral endocytosis. Here, we investigated the immunologic and virologic efficacy of baricitinib in a rhesus macaque model of SARS-CoV-2 infection. Viral shedding measured from nasal and throat swabs, bronchoalveolar lavages and tissues was not reduced with baricitinib. Type I IFN antiviral responses and SARS-CoV-2 specific T cell responses remained similar between the two groups. Importantly, however, animals treated with baricitinib showed reduced immune activation, decreased infiltration of neutrophils into the lung, reduced NETosis activity, and more limited lung pathology. Moreover, baricitinib treated animals had a rapid and remarkably potent suppression of alveolar macrophage derived production of cytokines and chemokines responsible for inflammation and neutrophil recruitment. These data support a beneficial role for, and elucidate the immunological mechanisms underlying, the use of baricitinib as a frontline treatment for severe inflammation induced by SARS-CoV-2 infection.
- Published
- 2020
- Full Text
- View/download PDF
9. Type I and Type III Interferons Restrict SARS-CoV-2 Infection of Human Airway Epithelial Cultures
- Author
-
Steven E. Bosinger, Shamika Bedoya, Philipp Ralfs, Kathryn L. Pellegrini, Arash Grakoui, Anice C. Lowen, Eric J. Sorscher, Bernardo A. Mainou, Hadj S. Aoued, Amit A. Upadhyay, Larry J. Anderson, Jenna L. Lobby, Candela Manfredi, Tatiana Chirkova, Jacob E. Kohlmeier, Vineet D. Menachery, Abigail Vanderheiden, Gregory M. Tharp, Mehul S. Suthar, Pei Yong Shi, and Matthew G. Zimmerman
- Subjects
Chemokine ,viruses ,Cellular Response to Infection ,medicine.disease_cause ,Virus Replication ,Madin Darby Canine Kidney Cells ,Interferon Lambda ,0302 clinical medicine ,Interferon ,Chlorocebus aethiops ,Lung ,Cells, Cultured ,Coronavirus ,0303 health sciences ,biology ,030220 oncology & carcinogenesis ,Interferon Type I ,Cytokines ,Chemokines ,Coronavirus Infections ,medicine.drug ,Immunology ,Pneumonia, Viral ,Bronchi ,Microbiology ,Virus ,Proinflammatory cytokine ,Cell Line ,03 medical and health sciences ,Betacoronavirus ,Immune system ,Dogs ,Virology ,medicine ,Animals ,Humans ,Interleukin 8 ,Pandemics ,Vero Cells ,030304 developmental biology ,Innate immune system ,SARS-CoV-2 ,fungi ,COVID-19 ,Epithelial Cells ,respiratory tract diseases ,Insect Science ,biology.protein ,Interferons - Abstract
The newly emerged human coronavirus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has caused a pandemic of respiratory illness. Current evidence suggests that severe cases of SARS-CoV-2 are associated with a dysregulated immune response. However, little is known about how the innate immune system responds to SARS-CoV-2. In this study, we modeled SARS-CoV-2 infection using primary human airway epithelial (pHAE) cultures, which are maintained in an air-liquid interface. We found that SARS-CoV-2 infects and replicates in pHAE cultures and is directionally released on the apical, but not basolateral, surface. Transcriptional profiling studies found that infected pHAE cultures had a molecular signature dominated by proinflammatory cytokines and chemokine induction, including interleukin 6 (IL-6), tumor necrosis factor alpha (TNF-α), and CXCL8, and identified NF-κB and ATF-4 as key drivers of this proinflammatory cytokine response. Surprisingly, we observed a complete lack of a type I or III interferon (IFN) response to SARS-CoV-2 infection. However, pretreatment and posttreatment with type I and III IFNs significantly reduced virus replication in pHAE cultures that correlated with upregulation of antiviral effector genes. Combined, our findings demonstrate that SARS-CoV-2 does not trigger an IFN response but is sensitive to the effects of type I and III IFNs. Our studies demonstrate the utility of pHAE cultures to model SARS-CoV-2 infection and that both type I and III IFNs can serve as therapeutic options to treat COVID-19 patients. IMPORTANCE The current pandemic of respiratory illness, COVID-19, is caused by a recently emerged coronavirus named SARS-CoV-2. This virus infects airway and lung cells causing fever, dry cough, and shortness of breath. Severe cases of COVID-19 can result in lung damage, low blood oxygen levels, and even death. As there are currently no vaccines approved for use in humans, studies of the mechanisms of SARS-CoV-2 infection are urgently needed. Our research identifies an excellent system to model SARS-CoV-2 infection of the human airways that can be used to test various treatments. Analysis of infection in this model system found that human airway epithelial cell cultures induce a strong proinflammatory cytokine response yet block the production of type I and III IFNs to SARS-CoV-2. However, treatment of airway cultures with the immune molecules type I or type III interferon (IFN) was able to inhibit SARS-CoV-2 infection. Thus, our model system identified type I or type III IFN as potential antiviral treatments for COVID-19 patients.
- Published
- 2020
10. Baricitinib treatment resolves lower-airway macrophage inflammation and neutrophil recruitment in SARS-CoV-2-infected rhesus macaques
- Author
-
Zachary Strongin, Timothy N. Hoang, Rebecca D. Levit, Shelly Wang, Sherrie Jean, Kimberly A. Cooney, Susan Pereira Ribeiro, Mirko Paiardini, Lanfang Wang, Gregory K. Tharp, Rafick Pierre Sekaly, Michael Nekorchuk, Elise G. Viox, Raymond F. Schinazi, Justin L. Harper, Jacob D. Estes, Steven E. Bosinger, Michelle Y.H. Lee, Hilmi Al-Shakhshir, Rachelle L. Stammen, Kathleen Busman-Sahay, Arun K. Boddapati, Daniela Weiskopf, Sijia Tao, Anne Piantadosi, Guido Silvestri, Kathryn L. Pellegrini, Maria Pino, Michael N. Sayegh, Fawn Connor-Stroud, Thomas H. Vanderford, Tristan R. Horton, Jesse J. Waggoner, Joyce Cohen, Olivia M. Delmas, Jennifer S. Wood, Peter D. Filev, Carly E. Starke, Amit A. Upadhyay, Sudhir Pai Kasturi, Ernestine A. Mahar, Shannon Kirejczyk, Elizabeth N. Beagle, Sanjeev Gumber, and Keivan Zandi
- Subjects
Chemokine ,T-Lymphocytes ,Anti-Inflammatory Agents ,Lymphocyte Activation ,Virus Replication ,Severity of Illness Index ,Cell Degranulation ,Pathogenesis ,immunology ,0302 clinical medicine ,Interferon ,Macrophage ,baricitinib ,Lung ,0303 health sciences ,Sulfonamides ,Cell Death ,pathogenesis ,respiratory system ,Neutrophil Infiltration ,medicine.symptom ,medicine.drug ,Inflammation ,nonhuman primate ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Article ,immune activation ,03 medical and health sciences ,Macrophages, Alveolar ,medicine ,Animals ,Viral shedding ,030304 developmental biology ,Janus Kinases ,Innate immune system ,SARS-CoV-2 ,COVID-19 ,Macaca mulatta ,respiratory tract diseases ,COVID-19 Drug Treatment ,Respiratory pharmacology ,Disease Models, Animal ,Purines ,Immunology ,biology.protein ,Azetidines ,Pyrazoles ,pharmacology ,030217 neurology & neurosurgery - Abstract
SARS-CoV-2 induced hypercytokinemia and inflammation are critically associated with COVID-19 disease severity. Baricitinib, a clinically approved JAK1/2 inhibitor, is currently being investigated in COVID-19 clinical trials. Here, we investigated the immunologic and virologic efficacy of baricitinib in a rhesus macaque model of SARS-CoV-2 infection. Viral shedding measured from nasal and throat swabs, bronchoalveolar lavages and tissues was not reduced with baricitinib. Type-I IFN antiviral responses and SARS-CoV-2-specific T-cell responses remained similar between the two groups. Animals treated with baricitinib showed reduced inflammation, decreased lung infiltration of inflammatory cells, reduced NETosis activity, and more limited lung pathology. Importantly, baricitinib treated animals had a rapid and remarkably potent suppression of lung macrophages production of cytokines and chemokines responsible for inflammation and neutrophil recruitment. These data support a beneficial role for, and elucidate the immunological mechanisms underlying, the use of baricitinib as a frontline treatment for inflammation induced by SARS-CoV-2 infection., Highlights • SARS-CoV-2 infected RMs mimic signatures of inflammation seen in COVID-19 patients • Baricitinib suppresses production of pro-inflammatory cytokines in lung macrophages • Baricitinib limits recruitment of neutrophils to the lung and NETosis • Baricitinib preserves innate antiviral and SARS-CoV-2-specific T-cell responses, Using a rhesus macaque infection model it is shown that baricitinib treatment started early after infection effectively resolves inflammatory signatures in airway macrophages, with decreased lung pathology and neutrophil infiltration.
- Published
- 2020
11. Type I and Type III IFN Restrict SARS-CoV-2 Infection of Human Airway Epithelial Cultures
- Author
-
Anice C. Lowen, Larry J. Anderson, Hadj S. Aoued, Kathryn L. Pellegrini, Mehul S. Suthar, Tatiana Chirkova, Matthew G. Zimmerman, Steven E. Bosinger, Amit A. Upadhyay, Shamika Bedoya, Vineet D. Menachery, Abigail Vanderheiden, Arash Grakoui, Philipp Ralfs, and Gregory M. Tharp
- Subjects
Chemokine ,Innate immune system ,Viral replication ,Downregulation and upregulation ,Effector ,biology.protein ,Tumor necrosis factor alpha ,Interleukin 8 ,Biology ,Virology ,Transcription factor ,respiratory tract diseases - Abstract
The newly emerged human coronavirus, SARS-CoV-2, has caused a pandemic of respiratory illness. The innate immune response is critical for protection against Coronaviruses. However, little is known about the interplay between the innate immune system and SARS-CoV-2. Here, we modeled SARS-CoV-2 infection using primary human airway epithelial (pHAE) cultures, which are maintained in an air-liquid interface. We found that SARS-CoV-2 infects and replicates in pHAE cultures and is directionally released on the apical, but not basolateral surface. Transcriptional profiling studies found that infected pHAE cultures had a molecular signature dominated by pro-inflammatory cytokines and chemokine induction, including IL-6, TNFα, CXCL8. We also identified NF-κB and ATF4 transcription factors as key drivers of this pro-inflammatory cytokine response. Surprisingly, we observed a complete lack of a type I or III IFN induction during SARS-CoV-2 infection. Pre-treatment or post-treatment with type I and III IFNs dramatically reduced virus replication in pHAE cultures and this corresponded with an upregulation of antiviral effector genes. Our findings demonstrate that SARS-CoV-2 induces a strong pro-inflammatory cytokine response yet blocks the production of type I and III IFNs. Further, SARS-CoV-2 is sensitive to the effects of type I and III IFNs, demonstrating their potential utility as therapeutic options to treat COVID-19 patients.IMPORTANCEThe current pandemic of respiratory illness, COVID-19, is caused by a recently emerged coronavirus named SARS-CoV-2. This virus infects airway and lung cells causing fever, dry cough, and shortness of breath. Severe cases of COVID-19 can result in lung damage, low blood oxygen levels, and even death. As there are currently no vaccines or antivirals approved for use in humans, studies of the mechanisms of SARS-CoV-2 infection are urgently needed. SARS-CoV-2 infection of primary human airway epithelial cultures induces a strong pro-inflammatory cytokine response yet blocks the production of type I and III IFNs. Further, SARS-CoV-2 is sensitive to the effects of type I and III IFNs, demonstrating their potential utility as therapeutic options to treat COVID-19 patients.
- Published
- 2020
- Full Text
- View/download PDF
12. Systems biological assessment of immunity to mild versus severe COVID-19 infection in humans
- Author
-
Steve E. Bosinger, Prabhu S. Arunachalam, Bali Pulendran, Amit A. Upadhyay, Thomas Hagan, Dmitri Kazmin, Thomas Shiu Hong Chik, Huibin Lv, Michele Paine McCullough, Srilatha Edupuganti, Purvesh Khatri, Ghina Alaaeddine, Nadine Rouphael, Dhananjay Wagh, Chris Ka Pun Mok, Evan J. Anderson, John A. Coller, Florian Wimmers, Natalia Sigal, Holden T. Maecker, Wai Shing Leung, Ranawaka A.P.M. Perera, Jacky Man Chun Chan, Madeleine K D Scott, Owen Tak-Yin Tsang, Christopher Huerta, Malik Peiris, Laurel Bristow, Yupeng Feng, Chris Yau Chung Choi, and Kathryn L. Pellegrini
- Subjects
DNA, Bacterial ,Lipopolysaccharides ,Male ,Transcription, Genetic ,Pneumonia, Viral ,Immunology ,Immunoglobulins ,Human leukocyte antigen ,Peripheral blood mononuclear cell ,Proinflammatory cytokine ,Transcriptome ,Betacoronavirus ,Immune system ,Immunity ,Medicine ,Humans ,Myeloid Cells ,Pandemics ,PI3K/AKT/mTOR pathway ,Research Articles ,Innate immune system ,Multidisciplinary ,business.industry ,SARS-CoV-2 ,Systems Biology ,TOR Serine-Threonine Kinases ,R-Articles ,COVID-19 ,Microbio ,Dendritic Cells ,HLA-DR Antigens ,Flow Cytometry ,Immunity, Innate ,Interferon Type I ,Leukocytes, Mononuclear ,Cytokines ,Female ,Inflammation Mediators ,Single-Cell Analysis ,business ,Coronavirus Infections ,Signal Transduction ,Research Article - Abstract
Immune profiling of COVID-19 patients Coronavirus disease 2019 (COVID-19) has affected millions of people globally, yet how the human immune system responds to and influences COVID-19 severity remains unclear. Mathew et al. present a comprehensive atlas of immune modulation associated with COVID-19. They performed high-dimensional flow cytometry of hospitalized COVID-19 patients and found three prominent and distinct immunotypes that are related to disease severity and clinical parameters. Arunachalam et al. report a systems biology approach to assess the immune system of COVID-19 patients with mild-to-severe disease. These studies provide a compendium of immune cell information and roadmaps for potential therapeutic interventions. Science, this issue p. eabc8511, p. 1210, Immune responses of COVID-19 patients are cataloged and compared with those of healthy individuals., Coronavirus disease 2019 (COVID-19) represents a global crisis, yet major knowledge gaps remain about human immunity to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). We analyzed immune responses in 76 COVID-19 patients and 69 healthy individuals from Hong Kong and Atlanta, Georgia, United States. In the peripheral blood mononuclear cells (PBMCs) of COVID-19 patients, we observed reduced expression of human leukocyte antigen class DR (HLA-DR) and proinflammatory cytokines by myeloid cells as well as impaired mammalian target of rapamycin (mTOR) signaling and interferon-α (IFN-α) production by plasmacytoid dendritic cells. By contrast, we detected enhanced plasma levels of inflammatory mediators—including EN-RAGE, TNFSF14, and oncostatin M—which correlated with disease severity and increased bacterial products in plasma. Single-cell transcriptomics revealed a lack of type I IFNs, reduced HLA-DR in the myeloid cells of patients with severe COVID-19, and transient expression of IFN-stimulated genes. This was consistent with bulk PBMC transcriptomics and transient, low IFN-α levels in plasma during infection. These results reveal mechanisms and potential therapeutic targets for COVID-19.
- Published
- 2020
13. Evaluation of a 24-gene signature for prognosis of metastatic events and prostate cancer-specific mortality
- Author
-
Qi Long, R. Jeffrey Karnes, Kasra Yousefi, Eric A. Klein, Martin G. Sanda, Robert B. Jenkins, Carlos S. Moreno, Kathryn L. Pellegrini, Elai Davicioni, Mandeep Takhar, Maria Santiago-Jimenez, Nicholas Erho, and Dattatraya Patil
- Subjects
Male ,Oncology ,medicine.medical_specialty ,Urology ,medicine.medical_treatment ,030232 urology & nephrology ,Disease-Free Survival ,Metastasis ,03 medical and health sciences ,Prostate cancer ,0302 clinical medicine ,Internal medicine ,medicine ,Humans ,Neoplasm Metastasis ,Survival analysis ,Prostatectomy ,Gynecology ,business.industry ,Prostatic Neoplasms ,Specific mortality ,Gene signature ,Microarray Analysis ,Prognosis ,medicine.disease ,030220 oncology & carcinogenesis ,Cohort ,Biomarker (medicine) ,Transcriptome ,business - Abstract
Objectives To determine the prognostic potential of Sig24 for identifying prostate cancer patients at risk of developing metastases or experiencing PCSM following radical prostatectomy. Subjects and methods Sig24 scores were calculated from previously collected gene expression microarray data from the Cleveland Clinic and Mayo Clinic (I and II). The performance of Sig24 was determined using time-dependent c-index analysis, Cox proportional hazards regression and Kaplan-Meier survival analysis. Results Higher Sig24 scores were significantly associated with higher pathologic Gleason scores (GS) in all three cohorts. Analysis of the Mayo Clinic II cohort, which included time to event information, indicated that patients with high Sig24 scores also had an increased risk of developing metastasis (HR: 3.78, 95% CI: 1.96-7.29, p < 0.001) or experiencing PCSM (HR: 6.54, 95% CI: 2.16-19.83, p < 0.001). Conclusions The findings of this study demonstrate the applicability of Sig24 for the prognosis of metastasis or PCSM following radical prostatectomy. Future studies investigating the combination of Sig24 with available prognostic tests may provide new approaches to improve risk stratification for patients with prostate cancer. This article is protected by copyright. All rights reserved.
- Published
- 2017
- Full Text
- View/download PDF
14. Predicting Immune Pathology after Hematopoietic Stem Cell Transplant with Transcriptomics: Naïve CD4 T Cell Expansion at Day 100 Predicts Patients with De Novo Chronic Gvhd
- Author
-
Amelia Langston, Yvonne Suessmuth, John T. Horan, Steven E. Bosinger, Victor Tkachev, Bruce R. Blazar, Kathryn L. Pellegrini, Muna Qayed, Kayla Betz, James Kaminski, Leslie S. Kean, Alison Yu, Ben Watkins, and Brandi Bratrude
- Subjects
Cd4 t cell ,business.industry ,Immunology ,Hematopoietic stem cell ,Cell Biology ,Hematology ,Biochemistry ,Transcriptome ,medicine.anatomical_structure ,Immune system ,immune system diseases ,hemic and lymphatic diseases ,Medicine ,Chronic gvhd ,business - Abstract
Background: Chronic graft-versus-host disease (CGVHD) is the leading cause of long-term morbidity and mortality following hematopoietic stem cell transplant (HCT) and occurs in over 50% of patients undergoing unrelated donor HCT. Despite its frequency, the mechanisms driving this disease remain incompletely understood, making its prevention and successful treatment challenging. To address this issue, we have undertaken a transcriptomic analysis of T cell reconstitution after unrelated donor HCT, to dissect mechanisms driving CGVHD. Methods: The patients studied were enrolled on a Phase 2, randomized, placebo-controlled trial of abatacept for GVHD prevention in patients receiving 8/8 unrelated-donor HCT for hematologic malignancies (NCT01743131). All immune analyses in the current study were performed on patients randomized to standard GVHD prophylaxis with calcineurin inhibition + methotrexate alone (placebo cohort, n =69), and thus provide insights into the drivers of CGVHD during standard unrelated donor HCT. On Day +100, CD4+ T cells were purified from the peripheral blood of these patients, and then analyzed by RNASeq. To determine the transcriptomic drivers of CGVHD without the confounder of significant prior acute GVHD (AGVHD) or exposure to steroids, we focused on profiling the CD4+ transcriptome of de novo CGVHD (CGVHD which develops in the absence of prior grade II-IV AGVHD, n = 7) and compared these patients to those who were 'operationally tolerant' and never developed either grade II-IV AGVHD or any CGVHD (n= 4). Gene expression from the resulting transcriptomes was quantified using kallisto. Differentially expressed (DE) genes were identified using DESeq2 (threshold for DE, adjusted (for multiple testing) p Results: DE analysis identified 101 genes that were significantly upregulated in CD4+ T cells from de novo CGVHD group and 54 genes that were significantly upregulated in the 'operationally tolerant' group (Figure 1A). GSEA identified that the mostly highly enriched signatures in patients with de novo CGVHD encompassed naïve CD4+ transcriptional programing (Figure 1B-C), in agreement with flow cytometric analysis, which also demonstrated expansion of CD4+ naïve T cells at Day +100 in patients developing de novo CGVHD compared to those demonstrating operational tolerance (Figure 1D). Importantly, the naïve CD4+ T cell signatures that were identified were distinct from those defining CD4+ stem cell memory T cells (which did not enrich in the de novo CGVHD cohort). In contrast, the gene signature of the operationally tolerant patients were enriched for regulatory gene sets (Figure 1C), consistent with a large body of evidence demonstrating that Treg expansion can be protective against CGVHD. Discussion: This study represents, to our knowledge, the first interrogation of the transcriptomic features of patients developing de novo CGVHD versus those operationally tolerant patients who develop neither significant AGVHD nor CGVHD after HCT. These patients may represent a particularly effective cohort in which to study immunologic drivers of CGVHD, given their freedom from prior treatment with corticosteroids, which can confound downstream transcriptomic analyses. Our data provide compelling evidence for a prominent naïve CD4+ T cell signature in patients who develop moderate-to-severe CGVHD despite their lack of antecedent AGVHD. These results are provocative, as they implicate a cell subset that is often considered more quiescent (naïve T cells) as associated with patients who develop immune pathology associated with CGVHD. These results suggest that naïve CD4+ T cells may represent a potent reservoir for alloreactivity, that, once activated, can cause significant disease. This would be in agreement with the implications of previously reported trials of naïve T cell depletion, which resulted in significant control of CGVHD. These results suggest that strategies to restrain naïve T cell pathogenic activation after Day +100 may improve CGVHD outcomes, and that the CD4+ T cell transcriptomic signature at this timepoint could be developed into a robust immunologic biomarker for the risk of developing CGVHD versus operational tolerance after HCT. Figure 1 Disclosures Watkins: Bristol Myers Squib: Honoraria. Qayed:Novartis: Consultancy; Mesoblast: Consultancy. Blazar:Tmunity: Other: Co-founder; KidsFirst Fund: Research Funding; BlueRock Therapeutics: Research Funding; Childrens' Cancer Research Fund: Research Funding; BlueRock Therapeuetic: Consultancy; Magenta Therapeutics: Consultancy; Fate Therapeutics Inc.: Research Funding. Horan:Bristol Myers Squib: Honoraria, Research Funding. Langston:Kadmon Corporation: Research Funding; Astellas Pharmaceuticals: Research Funding; Jazz Pharmaceuticals: Research Funding; Incyte: Research Funding; Bristol Myers Squib: Research Funding; Chimerix: Research Funding; Takeda: Research Funding. Kean:fortyseven: Consultancy; regeneron: Research Funding; hifibio: Consultancy; kymab: Consultancy; Bristol Meyers Squibb: Research Funding; gilead: Research Funding; novartis: Consultancy; bluebird bio: Research Funding; magenta: Research Funding.
- Published
- 2020
- Full Text
- View/download PDF
15. epiCaPture: A Urine DNA Methylation Test for Early Detection of Aggressive Prostate Cancer
- Author
-
Stephen P. Finn, Liam DeBarra, Robert D. Mills, Marcelino Yazbek Hanna, Bharati Bapat, Odharna Ni Dhomhnallain, William M. Gallagher, Julia Garcia, Rick Brugman, Alexandra V. Tuzova, Dattatraya Patil, Shane O’Meachair, Antoinette S. Perry, Connie M Dale, Elizabeth McEvoy, Fang Zhao, Rustom P. Manecksha, Anna L. Walsh, Olivia Schmidt, Gavin Clarke, Neil E. Fleshner, Eve O'Reilly, Niamh Russell, Jeremy Clark, Rachel Hurst, Kathryn L. Pellegrini, Martin G. Sanda, Odharnaith O'Brien, Colin Cooper, Sarah Kelly, and Daniel Brewer
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Prostate biopsy ,030232 urology & nephrology ,Article ,law.invention ,03 medical and health sciences ,Prostate cancer ,0302 clinical medicine ,law ,Prostate ,Internal medicine ,Medicine ,Epigenetics ,Prospective cohort study ,Polymerase chain reaction ,medicine.diagnostic_test ,business.industry ,Cancer ,medicine.disease ,6. Clean water ,3. Good health ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,DNA methylation ,business - Abstract
Purpose Liquid biopsies that noninvasively detect molecular correlates of aggressive prostate cancer (PCa) could be used to triage patients, reducing the burdens of unnecessary invasive prostate biopsy and enabling early detection of high-risk disease. DNA hypermethylation is among the earliest and most frequent aberrations in PCa. We investigated the accuracy of a six-gene DNA methylation panel (Epigenetic Cancer of the Prostate Test in Urine [epiCaPture]) at detecting PCa, high-grade (Gleason score greater than or equal to 8) and high-risk (D’Amico and Cancer of the Prostate Risk Assessment] PCa from urine. Patients and Methods Prognostic utility of epiCaPture genes was first validated in two independent prostate tissue cohorts. epiCaPture was assessed in a multicenter prospective study of 463 men undergoing prostate biopsy. epiCaPture was performed by quantitative methylation-specific polymerase chain reaction in DNA isolated from prebiopsy urine sediments and evaluated by receiver operating characteristic and decision curves (clinical benefit). The epiCaPture score was developed and validated on a two thirds training set to one third test set. Results Higher methylation of epiCaPture genes was significantly associated with increasing aggressiveness in PCa tissues. In urine, area under the receiver operating characteristic curve was 0.64, 0.86, and 0.83 for detecting PCa, high-grade PCa, and high-risk PCa, respectively. Decision curves revealed a net benefit across relevant threshold probabilities. Independent analysis of two epiCaPture genes in the same clinical cohort provided analytical validation. Parallel epiCaPture analysis in urine and matched biopsy cores showed added value of a liquid biopsy. Conclusion epiCaPture is a urine DNA methylation test for high-risk PCa. Its tumor specificity out-performs that of prostate-specific antigen (greater than 3 ng/mL). Used as an adjunct to prostate-specific antigen, epiCaPture could aid patient stratification to determine need for biopsy.
- Published
- 2019
16. Vascular Disease and Thrombosis in SARS-CoV-2-Infected Rhesus Macaques
- Author
-
Kathleen Busman-Sahay, Maria Pino, Jacob D. Estes, Maciel Porto, Hanne Leth Andersen, Steven E. Bosinger, Dan H. Barouch, Margaret Terry, Peter K. Sorger, Stephen Bondoc, Carly E. Starke, Zoltan Maliga, Linda M. Wrijil, Sarah Ducat, Kathryn E. Stephenson, Abishek Chandrashekar, Samuel J. Vidal, Malika Aid, Kathryn L. Pellegrini, Mark G. Lewis, Amanda J. Martinot, Timothy N. Hoang, Shivani A. Patel, Andrew D. Miller, Olga R. Brook, Jonathan L. Hecht, and Connor A. Jacobson
- Subjects
Male ,collagen ,Inflammation ,macrophage ,Biology ,Bronchoalveolar Lavage ,Article ,General Biochemistry, Genetics and Molecular Biology ,Proinflammatory cytokine ,Pathogenesis ,03 medical and health sciences ,0302 clinical medicine ,vascular ,medicine ,Animals ,Humans ,complement ,Platelet ,Vascular Diseases ,Platelet activation ,coagulation ,Complement Activation ,Lung ,thrombosis ,030304 developmental biology ,Aged, 80 and over ,platelet ,0303 health sciences ,medicine.diagnostic_test ,SARS-CoV-2 ,Vascular disease ,Macrophages ,COVID-19 ,Platelet Activation ,medicine.disease ,Macaca mulatta ,Thrombosis ,respiratory tract diseases ,C-Reactive Protein ,Bronchoalveolar lavage ,Immunology ,Cytokines ,Female ,vWF ,medicine.symptom ,Transcriptome ,IFNα ,030217 neurology & neurosurgery - Abstract
The COVID-19 pandemic has led to extensive morbidity and mortality throughout the world. Clinical features that drive SARS-CoV-2 pathogenesis in humans include inflammation and thrombosis, but the mechanistic details underlying these processes remain to be determined. In this study, we demonstrate endothelial disruption and vascular thrombosis in histopathologic sections of lungs from both humans and rhesus macaques infected with SARS-CoV-2. To define key molecular pathways associated with SARS-CoV-2 pathogenesis in macaques, we performed transcriptomic analyses of bronchoalveolar lavage and peripheral blood and proteomic analyses of serum. We observed macrophage infiltrates in lung and upregulation of macrophage, complement, platelet activation, thrombosis, and proinflammatory markers, including C-reactive protein, MX1, IL-6, IL-1, IL-8, TNFα, and NF-κB. These results suggest a model in which critical interactions between inflammatory and thrombosis pathways lead to SARS-CoV-2-induced vascular disease. Our findings suggest potential therapeutic targets for COVID-19., Graphical Abstract, Highlights • SARS-CoV-2 infection leads to macrophage infiltrates in the lung of infected macaques • SARS-CoV-2 upregulates proinflammatory cytokines and ISGs in macaques • SARS-CoV-2 upregulates complement and coagulation cascade in macaques • SARS-CoV-2 infection leads to endothelial damage and thrombosis in macaques, Aid et al. show that SARS-CoV-2 causes endothelial disruption and vascular thrombosis in both human and rhesus macaques lungs by inducing an upregulation of proinflammatory cytokines. Using an approach that combines histopathology and multiomics in macaques, they show the progression to vascular disease over time, which involves complement, macrophage, cytokine, and thrombosis cascades.
- Published
- 2020
- Full Text
- View/download PDF
17. MP35-10 DETECTION OF PROSTATE CANCER-ASSOCIATED TRANSCRIPTS IN URINARY EXTRACELLULAR VESICLES
- Author
-
Ella N Anastasiades, Eugene Huang, Frances Kim, Kristin Larsen, Kathryn L. Pellegrini, Carlos S. Moreno, Dattatraya Patil, Almira Catic, Kristen Douglas, Martin G. Sanda, and Mersiha Torlak
- Subjects
Prostate cancer ,business.industry ,Urology ,Urinary system ,Cancer research ,Medicine ,business ,medicine.disease ,Extracellular vesicles - Published
- 2018
- Full Text
- View/download PDF
18. Detection of prostate cancer-specific transcripts in extracellular vesicles isolated from post-DRE urine
- Author
-
Jeremy Clark, Kathryn Wehrmeyer, Kathryn L. Pellegrini, Mersiha Torlak, Martin G. Sanda, Colin Cooper, Kristen Douglas, Grace Lee, Dattatraya Patil, and Carlos S. Moreno
- Subjects
Adult ,Male ,0301 basic medicine ,PCA3 ,Pathology ,medicine.medical_specialty ,Urology ,Urine ,Urinalysis ,Biology ,Article ,Andrology ,Extracellular Vesicles ,03 medical and health sciences ,Prostate cancer ,0302 clinical medicine ,Transcriptional Regulator ERG ,Antigens, Neoplasm ,Prostate ,Gene expression ,Biomarkers, Tumor ,medicine ,Humans ,Biomarker discovery ,Early Detection of Cancer ,Aged ,Digital Rectal Examination ,medicine.diagnostic_test ,Gene Expression Profiling ,Prostatic Neoplasms ,Reproducibility of Results ,RNA ,Rectal examination ,Middle Aged ,medicine.disease ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis - Abstract
Background: The measurement of gene expression in post-digital rectal examination (DRE) urine specimens provides a non-invasive method to determine a patient's risk of prostate cancer. Many currently available assays use whole urine or cell pellets for the analysis of prostate cancer-associated genes, although the use of extracellular vesicles (EVs) has also recently been of interest. We investigated the expression of prostate-, kidney-, and bladder-specific transcripts and known prostate cancer biomarkers in urine EVs. Methods: Cell pellets and EVs were recovered from post-DRE urine specimens, with the total RNA yield and quality determined by Bioanalyzer. The levels of prostate, kidney, and bladder-associated transcripts in EVs were assessed by TaqMan qPCR and targeted sequencing. Results: RNA was more consistently recovered from the urine EV specimens, with over 80% of the patients demonstrating higher RNA yields in the EV fraction as compared to urine cell pellets. The median EV RNA yield of 36.4 ng was significantly higher than the median urine cell pellet RNA yield of 4.8 ng. Analysis of the post-DRE urine EVs indicated that prostate-specific transcripts were more abundant than kidney- or bladder-specific transcripts. Additionally, patients with prostate cancer had significantly higher levels of the prostate cancer-associated genes PCA3 and ERG. Conclusions: Post-DRE urine EVs are a viable source of prostate-derived RNAs for biomarker discovery and prostate cancer status can be distinguished from analysis of these specimens. Continued analysis of urine EVs offers the potential discovery of novel biomarkers for pre-biopsy prostate cancer detection.
- Published
- 2017
19. Androgens alter T-cell immunity by inhibiting T-helper 1 differentiation
- Author
-
Seung T. On, Laura Dunn, Martin G. Sanda, Haydn T. Kissick, Kathryn L. Pellegrini, Mohamed S. Arredouani, and Jonathan K. Noel
- Subjects
Male ,Cellular immunity ,medicine.drug_class ,T-Lymphocytes ,Cellular differentiation ,Biology ,urologic and male genital diseases ,Immune tolerance ,Androgen deprivation therapy ,Mice ,Prostate cancer ,Immune system ,medicine ,Animals ,Testosterone ,Phosphorylation ,Lung ,Protein Tyrosine Phosphatase, Non-Receptor Type 1 ,TYK2 Kinase ,Multidisciplinary ,Prostate ,Cell Differentiation ,Biological Sciences ,STAT4 Transcription Factor ,Th1 Cells ,Androgen ,medicine.disease ,Interleukin-12 ,Introns ,Up-Regulation ,Intestines ,Androgen receptor ,Immunology ,Cancer research ,Orchiectomy - Abstract
The hormonal milieu influences immune tolerance and the immune response against viruses and cancer, but the direct effect of androgens on cellular immunity remains largely uncharacterized. We therefore sought to evaluate the effect of androgens on murine and human T cells in vivo and in vitro. We found that murine androgen deprivation in vivo elicited RNA expression patterns conducive to IFN signaling and T-cell differentiation. Interrogation of mechanism showed that testosterone regulates T-helper 1 (Th1) differentiation by inhibiting IL-12-induced Stat4 phosphorylation: in murine models, we determined that androgen receptor binds a conserved region within the phosphatase, Ptpn1, and consequent up-regulation of Ptpn1 then inhibits IL-12 signaling in CD4 T cells. The clinical relevance of this mechanism, whereby the androgen milieu modulates CD4 T-cell differentiation, was ascertained as we found that androgen deprivation reduced expression of Ptpn1 in CD4 cells from patients undergoing androgen deprivation therapy for prostate cancer. Our findings, which demonstrate a clinically relevant mechanism by which androgens inhibit Th1 differentiation of CD4 T cells, provide rationale for targeting androgens to enhance CD4-mediated immune responses in cancer or, conversely, for modulating androgens to mitigate CD4 responses in disorders of autoimmunity.
- Published
- 2014
- Full Text
- View/download PDF
20. Transcriptomic Analysis of CD4+ T Cell Dysfunction during Gvhd: Evidence for Profound Reprograming of T Cell Signaling during Acute Gvhd That Is Controlled during CD28:CD80/86 Costimulation Blockade with Abatacept
- Author
-
Kayla Betz, Leslie S. Kean, Alison Yu, John T. Horan, Kathryn L. Pellegrini, Amelia Langston, Muna Qayed, Steven E. Bosinger, Yvonne Suessmuth, Benjamin Watkins, Bruce R. Blazar, and Brandi Bratrude
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Abatacept ,T cell ,medicine.medical_treatment ,Immunology ,Cell Biology ,Hematology ,Hematopoietic stem cell transplantation ,medicine.disease ,Biochemistry ,Transplantation ,Calcineurin ,medicine.anatomical_structure ,Graft-versus-host disease ,Internal medicine ,Lymphocyte costimulation ,Medicine ,business ,CD80 ,medicine.drug - Abstract
Although acute graft-versus-host-disease (AGVHD) is one of the major causes of non-relapse mortality after hematopoietic stem cell transplant (HCT), we are still unable to predict which patients will develop the most severe form of this disease, or which molecular pathways are dysregulated in the T cells that cause disease. Thus, understanding the molecular features of AGVHD is a critical unmet need. To address this, we have performed a companion mechanistic study as a part of our completed Phase 2 trial of abatacept, a CD28:CD80/86 costimulation blockade agent, for severe AGVHD prevention (Clinicaltrials.gov # NCT01743131, 'ABA2'). ABA2 has demonstrated significant improvement in AGVHD in patients prophylaxed with abatacept in addition to calcineurin inhibition (CNI) + Methotrexate (MTX) compared to controls receiving CNI/MTX alone. To begin to uncover mechanisms responsible for the control of AGVHD with abatacept, and given that CD4+ T cells have been consistently documented to be the main therapeutic target of this drug, we interrogated the transcriptome of CD4+ T cells reconstituting in patients prophylaxed with abatacept compared to CNI/MTX. To perform this analysis, we flow cytometrically sorted CD4+ T cells on Days 21-28 post-transplant from all patients on ABA2, as well as a cohort of 12 untransplanted healthy controls, and subsequently performed mRNA-sequencing on these cells. Weighted Gene Correlation Network Analysis (WGCNA) was performed on the top 6000 most variant transcripts from the resulting sequencing data. Hierarchical clustering of the WGCNA co-expression matrix enabled the identification of self-assembling modules (SAMs) that met a threshold of coexpression (Figure 1A). For the ABA2 dataset, we considered the following variables in the WGCNA model: patient cohort (7/8 patients, 8/8 patients, healthy controls), +/- prophylaxis with abatacept, CMV reactivation, EBV reactivation, Grade of GVHD (0-4), relapse, non-relapse mortality, and all-cause mortality. The WGCNA clustering analysis resulted in the identification of 4 discrete SAMs, which were highly correlated with clinical variable metamodules. This analysis revealed a strong positive correlation of a 476-gene SAM (the Turquoise module) in patients prophylaxed with CNI/MTX + placebo and anti-correlation of this module in patients prophylaxed with CNI/MTX + abatacept, as demonstrated in both the WGCNA heatmap and through Gene Set Enrichment Analysis (Figure 1 A-B). These opposing correlations suggested that interrogation of this module would reveal mechanistic correlates with standard prophylaxis that were decoupled by abatacept. Pathway analysis using the Reactome database (Figure 1C) revealed the turquoise SAM to be dominated by four types of pathways: (1) Those that define canonical cell-cycle pathways (2) Those involved in T cell metabolism (3) Those involved in apoptosis and (4) Those involved in T cell activation, consistent with upregulation of these transcripts in placebo versus abatacept patients. In addition to being highly correlated with patients receiving placebo, the expression of a subset of the transcripts in the Turquoise module were also directly correlated with the severity of AGVHD in these patients. Thus, linear regression analysis of the 476 transcripts in this module identified a subset of 93 genes for which transcript expression level was increased both in placebo compared to abatacept, and for which expression level also positively correlated with Grade of AGVHD. As with the Turquoise module as a whole, this subset of genes also formed a highly correlated network, linking transcripts involved in T cell proliferation, apoptosis, activation, metabolism as well as the T cell checkpoint (Figure 1D). This analysis represents the first comprehensive interrogation of the transcriptomic correlates of AGVHD. It identifies a novel set of transcripts which positively associate with the severity of AGVHD, and which costimulation blockade with abatacept down-regulates and de-couples from AGVHD severity. These results suggest a profound reprograming of T cell activation with abatacept that is correlated with the control of AGVHD. Disclosures Qayed: Bristol-Myers Squibb: Honoraria. Langston:Astellas Pharma: Other: Research Support; Incyte: Other: Research Support; Jazz Pharmaceuticals: Other: Research Support; Chimerix: Other: Research Support; Takeda: Other: Research Support; Kadmon Corporation: Other: Research Support; Novartis: Other: Research Support; Bristol Myers Squibb: Other: Research Support. Blazar:Fate Therapeutics, Inc.: Research Funding; RXi Pharmaceuticals: Research Funding; Alpine Immune Sciences, Inc.: Research Funding; Abbvie Inc: Research Funding; Leukemia and Lymphoma Society: Research Funding; Childrens' Cancer Research Fund: Research Funding; KidsFirst Fund: Research Funding; Tmunity: Other: Co-Founder; BlueRock Therapeutics: Membership on an entity's Board of Directors or advisory committees; Kamon Pharmaceuticals, Inc: Membership on an entity's Board of Directors or advisory committees; Five Prime Therapeutics Inc: Co-Founder, Membership on an entity's Board of Directors or advisory committees; Regeneron Pharmaceuticals: Membership on an entity's Board of Directors or advisory committees; Magenta Therapeutics and BlueRock Therapeuetics: Membership on an entity's Board of Directors or advisory committees. Kean:HiFiBio: Consultancy; BlueBirdBio: Research Funding; Gilead: Research Funding; Regeneron: Research Funding; EMDSerono: Consultancy; FortySeven: Consultancy; Magenta: Research Funding; Kymab: Consultancy; Jazz: Research Funding; Bristol Meyers Squibb: Patents & Royalties, Research Funding. OffLabel Disclosure: Abatacept: Approved for Rheumatoid Arthritis; used in this trial for prevention of GVHD.
- Published
- 2019
- Full Text
- View/download PDF
21. RNA-binding Protein Musashi Homologue 1 Regulates Kidney Fibrosis by Translational Inhibition of p21 and Numb mRNA*
- Author
-
Kathryn L. Pellegrini, Florin L. Craciun, Vishal S. Vaidya, Jenifer Seematti, Shreyas Jadhav, Amrendra Kumar Ajay, and Priyanka Trivedi
- Subjects
0301 basic medicine ,RNA-binding protein ,Nerve Tissue Proteins ,Biology ,Oncogene Protein p21(ras) ,Biochemistry ,03 medical and health sciences ,Mice ,Fibrosis ,medicine ,Animals ,Humans ,RNA, Messenger ,Molecular Biology ,Gene knockdown ,Kidney ,urogenital system ,HEK 293 cells ,RNA-Binding Proteins ,Cell Biology ,Cell cycle ,medicine.disease ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,HEK293 Cells ,Protein Biosynthesis ,NUMB ,RNA ,Kidney Diseases ,Kidney disease - Abstract
RNA-binding proteins (RBPs) are recognized as key posttranscriptional regulators that not only modulate the spatiotemporal expression of genes during organism development but also regulate disease pathogenesis. Very limited information exists on the potential role of RBPs in modulating kidney fibrosis, which is a major hallmark of chronic kidney disease. Here, we report a novel mechanism in kidney fibrosis involving a RBP, Musashi homologue 1 (Msi1), which is expressed in tubular epithelial cells. Using two mechanistically distinct mouse models of kidney fibrosis, we show that Msi1 protein levels are significantly down-regulated in the kidneys following fibrosis. We found that Msi1 functions by negatively regulating the translation of its target mRNAs, p21 and Numb, whose protein levels are markedly increased in kidney fibrosis. Also, Msi1 overexpression and knockdown in kidney epithelial cells cause p21- and Numb-mediated cell cycle arrest. Furthermore, we observed that Numb looses its characteristic membrane localization in fibrotic kidneys and therefore is likely unable to inhibit Notch resulting in tubular cell death. Oleic acid is a known inhibitor of Msi1 and injecting oleic acid followed by unilateral ureteral obstruction surgery in mice resulted in enhanced fibrosis compared with the control group, indicating that inhibiting Msi1 activity renders the mice more susceptible to fibrosis. Given that deregulated fatty acid metabolism plays a key role in kidney fibrosis, these results demonstrate a novel connection between fatty acid and Msi1, an RNA-binding protein, in kidney fibrosis.
- Published
- 2016
22. MP02-09 TARGETED SEQUENCING OF PROSTATE CANCER-ASSOCIATED RNAS IN EXTRACELLULAR VESICLES FROM POST-DRE URINE
- Author
-
Martin G. Sanda, Kathryn Wehrmeyer, Anna Bausum, Kathryn L. Pellegrini, Mersiha Torlak, Kristen Douglas, Carlos S. Moreno, and Dattatraya Patil
- Subjects
Prostate cancer ,business.industry ,Urology ,Medicine ,Urine ,business ,medicine.disease ,Extracellular vesicles ,Molecular biology - Published
- 2016
- Full Text
- View/download PDF
23. Identification of the Transcription Factor Relationships Associated with Androgen Deprivation Therapy Response and Metastatic Progression in Prostate Cancer
- Author
-
Michael R. Freeman, Sungyong You, Nitya V. Sharma, Kenneth Watanabe, Karen N. Conneely, Carlos S. Moreno, Adeboye O. Osunkoya, Felipe O. Giuste, Fred Saad, S.S. Ramalingam, Anne-Marie Mes-Masson, Dominique Trudel, Veronique Ouellet, John A. Petros, Paula M. Vertino, Lucresse Fossouo, Kathryn L. Pellegrini, and Eloise Adam-Granger
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,JUNB ,medicine.medical_treatment ,androgen deprivation therapy ,lcsh:RC254-282 ,Article ,Metastasis ,Transcriptome ,Androgen deprivation therapy ,03 medical and health sciences ,SOX4 ,Prostate cancer ,transcriptional networks ,Internal medicine ,medicine ,metastasis ,Prostatectomy ,business.industry ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,prostate cancer ,medicine.disease ,3. Good health ,030104 developmental biology ,business ,FOSB - Abstract
Background: Patients with locally advanced or recurrent prostate cancer typically undergo androgen deprivation therapy (ADT), but the benefits are often short-lived and the responses variable. ADT failure results in castration-resistant prostate cancer (CRPC), which inevitably leads to metastasis. We hypothesized that differences in tumor transcriptional programs may reflect differential responses to ADT and subsequent metastasis. Results: We performed whole transcriptome analysis of 20 patient-matched Pre-ADT biopsies and 20 Post-ADT prostatectomy specimens, and identified two subgroups of patients (high impact and low impact groups) that exhibited distinct transcriptional changes in response to ADT. We found that all patients lost the AR-dependent subtype (PCS2) transcriptional signatures. The high impact group maintained the more aggressive subtype (PCS1) signal, while the low impact group more resembled an AR-suppressed (PCS3) subtype. Computational analyses identified transcription factor coordinated groups (TFCGs) enriched in the high impact group network. Leveraging a large public dataset of over 800 metastatic and primary samples, we identified 33 TFCGs in common between the high impact group and metastatic lesions, including SOX4/FOXA2/GATA4, and a TFCG containing JUN, JUNB, JUND, FOS, FOSB, and FOSL1. The majority of metastatic TFCGs were subsets of larger TFCGs in the high impact group network, suggesting a refinement of critical TFCGs in prostate cancer progression. Conclusions: We have identified TFCGs associated with pronounced initial transcriptional response to ADT, aggressive signatures, and metastasis. Our findings suggest multiple new hypotheses that could lead to novel combination therapies to prevent the development of CRPC following ADT.
- Published
- 2018
- Full Text
- View/download PDF
24. Abstract 2269: Transcription factor relationships associated with androgen-deprivation therapy response and metastatic progression in prostate cancer
- Author
-
Paula M. Vertino, Veronique Ouellet, Kathryn L. Pellegrini, Adeboye O. Osunkoya, Eloise Adam-Granger, Kenneth Watanabe, Fred Saad, Anne-Marie Mes-Masson, Sungyong You, Michael R. Freeman, Carlos S. Moreno, Lucresse Fossouo, Nitya V. Sharma, Karen N. Conneely, John A. Petros, S.S. Ramalingam, Dominique Trudel, and Felipe O. Giuste
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,JUNB ,business.industry ,Cancer ,medicine.disease ,Androgen deprivation therapy ,Androgen receptor ,Prostate cancer ,SOX4 ,Internal medicine ,GLI2 ,medicine ,business ,Transcription factor - Abstract
Patients with recurrent, aggressive prostate cancer typically undergo androgen-deprivation therapy (ADT), but the benefits are often short-lived, and responses are variable. Failure to respond to ADT invariably leads to metastatic disease, and ultimately death. To investigate differential responses to ADT, we performed whole-transcriptome analysis of 20 patient-matched pre-ADT biopsies and post-ADT prostatectomy specimens, and observed that all patients lost transcriptional signatures indicative of the androgen receptor (AR)-dependent subtype, after treatment. We also identified two subgroups of patients with either a strong or weak transcriptional response to ADT. The strong responders maintained the more aggressive subtype signal, while the weak responders lost expression of these genes and more resembled an AR-suppressed, basal subtype. We generated a strong responder transcriptional network using the PANDA program and integrated expression data from our cohort, protein-protein interaction, and DNA binding motif data. We also leveraged the expression data from a large public dataset of over 800 metastatic and primary samples to construct a metastatic lesion transcriptional network. We identified 20 common transcription factor coordinated groups (TFCGs) associated with both the strong responders and metastatic lesions, including GLI3/GLI2, SOX4/FOXA2/GATA4, ERF/ETV5/ETV3/ELF4, and a TFCG containing JUN, JUNB, JUND, FOS, FOSB, and FOSL1. Many TFCGs in the metastatic network were subsets of larger groups in the strong responders network, implicating these transcription factor associations as potentially critical for both the differential ADT response and metastatic disease progression. Citation Format: Nitya V. Sharma, Kathryn L. Pellegrini, Veronique Ouellet, Felipe O. Giuste, Selvi Ramalingam, Kenneth Watanabe, Eloise Adam-Granger, Lucresse Fossouo, Sungyong You, Michael R. Freeman, Paula Vertino, Karen Conneely, Adeboye O. Osunkoya, Dominique Trudel, Anne-Marie Mes-Masson, John A. Petros, Fred Saad, Carlos S. Moreno. Transcription factor relationships associated with androgen-deprivation therapy response and metastatic progression in prostate cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 2269.
- Published
- 2018
- Full Text
- View/download PDF
25. Application of Small RNA Sequencing to Identify MicroRNAs in Acute Kidney Injury and Fibrosis
- Author
-
Haydn T. Kissick, Vanesa Bijol, Florin L. Craciun, Cory V. Gerlach, Kathryn L. Pellegrini, Vishal S. Vaidya, and Krithika Ramachandran
- Subjects
0301 basic medicine ,Male ,Small RNA ,Pathology ,medicine.medical_specialty ,Aristolochic acid ,In situ hybridization ,Biology ,Toxicology ,Article ,Kidney Tubules, Proximal ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,Folic Acid ,Fibrosis ,microRNA ,medicine ,Animals ,In Situ Hybridization ,Pharmacology ,Mice, Inbred BALB C ,Acute kidney injury ,RNA ,Acute Kidney Injury ,medicine.disease ,MicroRNAs ,030104 developmental biology ,chemistry ,Reperfusion injury - Abstract
Establishing a microRNA (miRNA) expression profile in affected tissues provides an important foundation for the discovery of miRNAs involved in the development or progression of pathologic conditions. We conducted small RNA sequencing to generate a temporal profile of miRNA expression in the kidneys using a mouse model of folic acid-induced (250 mg/kg i.p.) kidney injury and fibrosis. From the 103 miRNAs that were differentially expressed over the time course (>2-fold, p
- Published
- 2015
26. The transcription factor ERG increases expression of neurotransmitter receptors on prostate cancer cells
- Author
-
Haydn T. Kissick, Martin G. Sanda, John M. Asara, Seung T. On, Laura Dunn, Kathryn L. Pellegrini, Jonathan K. Noel, and Mohamed S. Arredouani
- Subjects
Male ,Nicotine ,Cancer Research ,medicine.medical_specialty ,Oncogene Proteins, Fusion ,genetic structures ,Receptors, Nicotinic ,urologic and male genital diseases ,Fusion gene ,03 medical and health sciences ,0302 clinical medicine ,Transcriptional Regulator ERG ,Neurotransmitter receptor ,Cell Line, Tumor ,Internal medicine ,LNCaP ,Genetics ,medicine ,Humans ,Receptor ,Transcription factor ,Cell Proliferation ,030304 developmental biology ,0303 health sciences ,business.industry ,Smoking ,Prostatic Neoplasms ,Transfection ,eye diseases ,Up-Regulation ,3. Good health ,Endocrinology ,Oncology ,030220 oncology & carcinogenesis ,Metabolome ,Trans-Activators ,Cancer research ,Calcium ,sense organs ,business ,Erg ,Research Article - Abstract
Background The TMPRSS2-ERG gene fusion occurs in about half of prostate cancer (PCa) cases and results in overexpression of the transcription factor ERG. Overexpression of ERG has many effects on cellular function. However, how these changes enhance cell growth and promote tumor development is unclear. Methods To investigate the role of ERG, LNCaP and PC3 cells were transfected with ERG and gene expression and metabolic profile were analyzed. Results Our data show that expression of ERG induces overexpression of many nicotinicacetylcholine receptors (nAChRs). In addition, metabolic profiling by LC-MS/MS revealed elevated production of several neurotransmitters in cells expressing ERG. Consistently, treatment of ERG-expressing cells with nicotine induced elevated calcium influx, GSK3β (Ser9) phosphorylation and cell proliferation. Finally, we show that PCa patientswho are smokers have larger tumors if their tumors are TMPRSS2-ERG gene fusion positive. Conclusion Collectively, our data suggest that ERG sensitizes prostate tumor cells to neurotransmitter receptor agonists like nicotine. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1612-3) contains supplementary material, which is available to authorized users.
- Published
- 2015
- Full Text
- View/download PDF
27. RNA biomarkers to facilitate the identification of aggressive prostate cancer
- Author
-
Carlos S. Moreno, Kathryn L. Pellegrini, and Martin G. Sanda
- Subjects
Male ,RNA, Untranslated ,Clinical Biochemistry ,Bioinformatics ,Biochemistry ,Article ,Fusion gene ,Prostate cancer ,Biopsy ,medicine ,Biomarkers, Tumor ,Humans ,RNA, Messenger ,Biomarker discovery ,Molecular Biology ,Gene Rearrangement ,medicine.diagnostic_test ,business.industry ,RNA ,Prostatic Neoplasms ,General Medicine ,medicine.disease ,Non-coding RNA ,Prognosis ,Gene Expression Regulation, Neoplastic ,Molecular Medicine ,Biomarker (medicine) ,Identification (biology) ,business - Abstract
A large number of men are diagnosed with prostate cancer each year, but many will not experience morbidity or mortality as a result of their cancers. Therefore, biomarkers for prostate cancer are necessary to carefully select patients for initial diagnostic biopsy or to facilitate care decisions for men who have already been diagnosed with prostate cancer. RNA-based approaches to biomarker discovery allow the investigation of non-coding RNAs, gene fusion transcripts, splice variants, and multi-gene expression panels in tissue, urine, or blood as opportunities to improve care decisions. This review focuses on RNA biomarkers that are available as commercial assays, and therefore already available for potential clinical use, as well as providing an overview of newer RNA biomarkers that are in earlier stages of clinical development.
- Published
- 2015
28. MicroRNA-155 deficient mice experience heightened kidney toxicity when dosed with cisplatin
- Author
-
William W. Chen, Kathryn L. Pellegrini, Florin L. Craciun, Vanesa Bijol, James C. Fuscoe, Vishal S. Vaidya, Janani Saikumar, and Tao Han
- Subjects
Male ,Programmed cell death ,Time Factors ,Apoptosis ,Biology ,Pharmacology ,Toxicology ,medicine.disease_cause ,Kidney ,Nephrotoxicity ,medicine ,Animals ,Cisplatin ,Mice, Knockout ,Gene Expression Profiling ,Computational Biology ,Acute Kidney Injury ,Fibrosis ,Up-Regulation ,Mice, Inbred C57BL ,Disease Models, Animal ,MicroRNAs ,Oxidative Stress ,medicine.anatomical_structure ,Toxic injury ,Immunology ,Toxicity ,Proto-Oncogene Proteins c-fos ,Oxidative stress ,medicine.drug ,Signal Transduction - Abstract
The development of nephrotoxicity limits the maximum achievable dosage and treatment intervals for cisplatin chemotherapy. Therefore, identifying mechanisms that regulate this toxicity could offer novel methods to optimize cisplatin delivery. MicroRNAs are capable of regulating many different genes, and can influence diverse cellular processes, including cell death and apoptosis. We previously observed miR-155 to be highly increased following ischemic or toxic injury to the kidneys and, therefore, sought to determine whether mice deficient in miR-155 would respond differently to kidney injury. We treated C57BL/6 and miR-155(-/-) mice with 20 mg/kg of cisplatin and found a significantly higher level of kidney injury in the miR-155(-/-) mice. Genome-wide expression profiling and bioinformatic analysis indicated the activation of a number of canonical signaling pathways relating to apoptosis and oxidative stress over the course of the injury, and identified potential upstream regulators of these effects. One predicted upstream regulator was c-Fos, which has two confirmed miR-155 binding sites in its 3' UTR and, therefore, can be directly regulated by miR-155. We established that the miR-155(-/-) mice had significantly higher levels of c-Fos mRNA and protein than the C57BL/6 mice at 72 h after cisplatin exposure. These data indicate a role for miR-155 in the cisplatin response and suggest that targeting of c-Fos could be investigated to reduce cisplatin-induced nephrotoxicity.
- Published
- 2014
29. The survival of myoblasts after intramuscular transplantation is improved when fewer cells are injected
- Author
-
Kathryn L. Pellegrini and Manfred W. Beilharz
- Subjects
Male ,medicine.medical_specialty ,Donor cell ,Cell Survival ,Cell Transplantation ,Duchenne muscular dystrophy ,Myoblasts, Skeletal ,Cell ,Hindlimb ,Injections, Intramuscular ,Andrology ,Mice ,Medicine ,Ischemic necrosis ,Myocyte ,Animals ,Muscle, Skeletal ,Transplantation ,biology ,business.industry ,medicine.disease ,Surgery ,Mice, Inbred C57BL ,Muscular Dystrophy, Duchenne ,Disease Models, Animal ,medicine.anatomical_structure ,biology.protein ,Mice, Inbred mdx ,Female ,business ,Dystrophin - Abstract
Background. Myoblast transplantation has long been studied as a potential therapy for Duchenne muscular dystrophy as the incorporation of donor myoblasts into host muscle allows the production of functional dystrophin protein. However, the clinical feasibility of this approach is limited by the poor survival of the donor cells in the weeks after transplantation. It has recently been determined that the intramuscular transplantation of large numbers of cells can lead to the formation of ischemic necrosis in the center of these cell masses. For this reason, the relationship between donor cell survival and the number of cells transplanted was investigated. Methods. Myoblasts were prepared from the hind limb muscles of male C57BL/10Sn mice and transplanted into the tibialis anterior muscles of female mdx mice at one of the following amounts: 103, 104, 105, or 106 cells. The survival of the transplanted cells was analyzed using a Y chromosome-specific qPCR. Results. It was found that donor cell survival was improved 1 week after transplantation when fewer myoblasts were transplanted, including the observation of donor cell proliferation after the transplantation of 103 myoblasts. However, concentration effects and long-term survival complicate the interpretation of these results. Conclusions. These results indicate that early donor myoblast survival was dependent on the number of cells transplanted and the volume of liquid used to deliver them into the muscle. We believe that this finding has implications for the design and interpretation of future experimentation relating to intramuscular cell therapies.
- Published
- 2010
30. Nanoparticle Detection of Urinary Markers for Point-of-Care Diagnosis of Kidney Injury
- Author
-
Kyungheon Lee, Hyun Jung Chung, Vishal S. Vaidya, Jaehoon Chung, Ralph Weissleder, Kathryn L. Pellegrini, Kamani Wanigasuriya, Hakho Lee, and Innocent Jayawardene
- Subjects
Adult ,medicine.medical_specialty ,Pathology ,Magnetic Resonance Spectroscopy ,Adolescent ,Point-of-Care Systems ,Urinary system ,Urology ,lcsh:Medicine ,Urine ,Sensitivity and Specificity ,Cohort Studies ,Young Adult ,chemistry.chemical_compound ,Microchip Analytical Procedures ,Kidney injury ,Humans ,Medicine ,Hepatitis A Virus Cellular Receptor 1 ,Cystatin C ,Renal Insufficiency, Chronic ,Magnetite Nanoparticles ,lcsh:Science ,Aged ,Point of care ,Aged, 80 and over ,Creatinine ,Membrane Glycoproteins ,Multidisciplinary ,biology ,business.industry ,lcsh:R ,Acute kidney injury ,Reproducibility of Results ,Heavy metals ,Acute Kidney Injury ,Middle Aged ,medicine.disease ,3. Good health ,chemistry ,biology.protein ,Feasibility Studies ,Receptors, Virus ,lcsh:Q ,business ,Biomarkers ,Research Article - Abstract
The high incidence of acute and chronic kidney injury due to various environmental factors such as heavy metals or chemicals has been a major problem in developing countries. However, the diagnosis of kidney injury in these areas can be more challenging due to the lack of highly sensitive and specific techniques that can be applied in point-of-care settings. To address this, we have developed a technique called ‘micro-urine nanoparticle detection (μUNPD)’, that allows the detection of trace amounts of molecular markers in urine. Specifically, this technique utilizes an automated on-chip assay followed by detection with a hand-held device for the read-out. Using the μUNPD technology, the kidney injury markers KIM-1 and Cystatin C were detected down to concentrations of 0.1 ng/ml and 20 ng/ml respectively, which meets the cut-off range required to identify patients with acute or chronic kidney injury. Thus, we show that the μUNPD technology enables point of care and non-invasive detection of kidney injury, and has potential for applications in diagnosing kidney injury with high sensitivity in resource-limited settings.
- Published
- 2015
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.