Your browser doesn't support javascript.
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Adicionar filtros

Tipo de documento
Intervalo de ano
1.
medrxiv; 2024.
Preprint em Inglês | medRxiv | ID: ppzbmed-10.1101.2024.02.05.24301794

RESUMO

Pregnancy is a risk factor for increased severity of SARS-CoV-2 and other respiratory infections. The mechanisms underlying this risk have not been well-established, partly due to a limited understanding of how pregnancy shapes immune responses. To gain insight into the role of pregnancy in modulating immune responses at steady state and upon perturbation, we collected peripheral blood mononuclear cells (PBMC), plasma, and stool from 226 women, including 152 pregnant individuals (n = 96 with SARS-CoV-2 infection and n = 56 healthy controls) and 74 non-pregnant women (n = 55 with SARS-CoV-2 and n = 19 healthy controls). We found that SARS-CoV-2 infection was associated with altered T cell responses in pregnant compared to non-pregnant women. Differences included a lower percentage of memory T cells, a distinct clonal expansion of CD4-expressing CD8+ T cells, and the enhanced expression of T cell exhaustion markers, such as programmed cell death-1 (PD-1) and T cell immunoglobulin and mucin domain-3 (Tim-3), in pregnant women. We identified additional evidence of immune dysfunction in severely and critically ill pregnant women, including a lack of expected elevation in regulatory T cell (Treg) levels, diminished interferon responses, and profound suppression of monocyte function. Consistent with earlier data, we found maternal obesity was also associated with altered immune responses to SARS-CoV-2 infection, including enhanced production of inflammatory cytokines by T cells. Certain gut bacterial species were altered in pregnancy and upon SARS-CoV-2 infection in pregnant individuals compared to non-pregnant women. Shifts in cytokine and chemokine levels were also identified in the sera of pregnant individuals, most notably a robust increase of interleukin-27 (IL-27), a cytokine known to drive T cell exhaustion, in the pregnant uninfected control group compared to all non-pregnant groups. IL-27 levels were also significantly higher in uninfected pregnant controls compared to pregnant SARS-CoV-2-infected individuals. Using two different preclinical mouse models of inflammation-induced fetal demise and respiratory influenza viral infection, we found that enhanced IL-27 protects developing fetuses from maternal inflammation but renders adult female mice vulnerable to viral infection. These combined findings from human and murine studies reveal nuanced pregnancy-associated immune responses, suggesting mechanisms underlying the increased susceptibility of pregnant individuals to viral respiratory infections.


Assuntos
Morte Fetal , Síndrome Respiratória Aguda Grave , Obesidade , Doenças do Sistema Imunitário , Infecções Respiratórias , Viroses , COVID-19 , Inflamação
2.
Toni M. Delorey; Carly G. K. Ziegler; Graham Heimberg; Rachelly Normand; Yiming Yang; Asa Segerstolpe; Domenic Abbondanza; Stephen J. Fleming; Ayshwarya Subramanian; Daniel T. Montoro; Karthik A. Jagadeesh; Kushal Dey; Pritha Sen; Michal Slyper; Yered Pita-Juarez; Devan Phillips; Zohar Bloom-Ackermann; Nick Barkas; Andrea Ganna; James Gomez; Erica Normandin; Pourya Naderi; Yury V. Popov; Siddharth S. Raju; Sebastian Niezen; Linus T.-Y. Tsai; Katherine J. Siddle; Malika Sud; Victoria M. Tran; Shamsudheen Karuthedath Vellarikkal; Liat Amir-Zilberstein; Joseph M Beechem; Olga R. Brook; Jonathan Chen; Prajan Divakar; Phylicia Dorceus; Jesse M Engreitz; Adam Essene; Donna M. Fitzgerald; Robin Fropf; Steven Gazal; Joshua Gould; Tyler Harvey; Jonathan Hecht; Tyler Hether; Judit Jane-Valbuena; Michael Leney-Greene; Hui Ma; Cristin McCabe; Daniel E. McLoughlin; Eric M. Miller; Christoph Muus; Mari Niemi; Robert Padera; Liuliu Pan; Deepti Pant; Jenna Pfiffner-Borges; Christopher J. Pinto; Jason Reeves; Marty Ross; Melissa Rudy; Erroll H. Rueckert; Michelle Siciliano; Alexander Sturm; Ellen Todres; Avinash Waghray; Sarah Warren; Shuting Zhang; Dan Zollinger; Lisa Cosimi; Rajat M Gupta; Nir Hacohen; Winston Hide; Alkes L. Price; Jayaraj Rajagopal; Purushothama Rao Tata; Stefan Riedel; Gyongyi Szabo; Timothy L. Tickle; Deborah Hung; Pardis C. Sabeti; Richard Novak; Robert Rogers; Donald E. Ingber; Z Gordon Jiang; Dejan Juric; Mehrtash Babadi; Samouil L. Farhi; James R. Stone; Ioannis S. Vlachos; Isaac H. Solomon; Orr Ashenberg; Caroline B.M. Porter; Bo Li; Alex K. Shalek; Alexandra-Chloe Villani; Orit Rozenblatt-Rosen; Aviv Regev.
biorxiv; 2021.
Preprint em Inglês | bioRxiv | ID: ppzbmed-10.1101.2021.02.25.430130

RESUMO

The SARS-CoV-2 pandemic has caused over 1 million deaths globally, mostly due to acute lung injury and acute respiratory distress syndrome, or direct complications resulting in multiple-organ failures. Little is known about the host tissue immune and cellular responses associated with COVID-19 infection, symptoms, and lethality. To address this, we collected tissues from 11 organs during the clinical autopsy of 17 individuals who succumbed to COVID-19, resulting in a tissue bank of approximately 420 specimens. We generated comprehensive cellular maps capturing COVID-19 biology related to patients demise through single-cell and single-nucleus RNA-Seq of lung, kidney, liver and heart tissues, and further contextualized our findings through spatial RNA profiling of distinct lung regions. We developed a computational framework that incorporates removal of ambient RNA and automated cell type annotation to facilitate comparison with other healthy and diseased tissue atlases. In the lung, we uncovered significantly altered transcriptional programs within the epithelial, immune, and stromal compartments and cell intrinsic changes in multiple cell types relative to lung tissue from healthy controls. We observed evidence of: alveolar type 2 (AT2) differentiation replacing depleted alveolar type 1 (AT1) lung epithelial cells, as previously seen in fibrosis; a concomitant increase in myofibroblasts reflective of defective tissue repair; and, putative TP63+ intrapulmonary basal-like progenitor (IPBLP) cells, similar to cells identified in H1N1 influenza, that may serve as an emergency cellular reserve for severely damaged alveoli. Together, these findings suggest the activation and failure of multiple avenues for regeneration of the epithelium in these terminal lungs. SARS-CoV-2 RNA reads were enriched in lung mononuclear phagocytic cells and endothelial cells, and these cells expressed distinct host response transcriptional programs. We corroborated the compositional and transcriptional changes in lung tissue through spatial analysis of RNA profiles in situ and distinguished unique tissue host responses between regions with and without viral RNA, and in COVID-19 donor tissues relative to healthy lung. Finally, we analyzed genetic regions implicated in COVID-19 GWAS with transcriptomic data to implicate specific cell types and genes associated with disease severity. Overall, our COVID-19 cell atlas is a foundational dataset to better understand the biological impact of SARS-CoV-2 infection across the human body and empowers the identification of new therapeutic interventions and prevention strategies.


Assuntos
Fibrose , Adenocarcinoma Bronquioloalveolar , Síndrome do Desconforto Respiratório , Lesão Pulmonar Aguda , COVID-19
3.
medrxiv; 2020.
Preprint em Inglês | medRxiv | ID: ppzbmed-10.1101.2020.07.21.20159376

RESUMO

IMPORTANCE: Microvascular lesions are common in patients with severe COVID-19. Radiologic-pathologic correlation in one case suggests a combination of microvascular hemorrhagic and ischemic lesions that may reflect an underlying hypoxic mechanism of injury, which requires validation in larger studies. OBJECTIVE: To determine the incidence, distribution, and clinical and histopathologic correlates of microvascular lesions in patients with severe COVID-19. DESIGN: Observational, retrospective cohort study: March to May 2020. SETTING: Single academic medical center. PARTICIPANTS: Consecutive patients (16) admitted to the intensive care unit with severe COVID-19, undergoing brain MRI for evaluation of coma or focal neurologic deficits. EXPOSURES: Not applicable. MAIN OUTCOME AND MEASURES: Hypointense microvascular lesions identified by a prototype ultrafast high-resolution susceptibility-weighted imaging (SWI) MRI sequence, counted by two neuroradiologists and categorized by neuroanatomic location. Clinical and laboratory data (most recent measurements before brain MRI). Brain autopsy and cerebrospinal fluid PCR for SARS-CoV 2 in one patient who died from severe COVID-19. RESULTS: Eleven of 16 patients (69%) had punctate and linear SWI lesions in the subcortical and deep white matter, and eight patients (50%) had >10 SWI lesions. In 4/16 patients (25%), lesions involved the corpus callosum. Brain autopsy in one patient revealed that SWI lesions corresponded to widespread microvascular injury, characterized by perivascular and parenchymal petechial hemorrhages and microscopic ischemic lesions. CONCLUSIONS AND RELEVANCE: SWI lesions are common in patients with neurological manifestations of severe COVID-19 (coma and focal neurologic deficits). The distribution of lesions is similar to that seen in patients with hypoxic respiratory failure, sepsis, and disseminated intravascular coagulation. Collectively, these radiologic and histopathologic findings suggest that patients with severe COVID-19 are at risk for multifocal microvascular hemorrhagic and ischemic lesions in the subcortical and deep white matter.


Assuntos
Hemorragia , Manifestações Neurológicas , Coagulação Intravascular Disseminada , Angina Microvascular , Síndrome Respiratória Aguda Grave , Sepse , Isquemia Encefálica , Coma , Icterícia Obstrutiva , COVID-19 , Insuficiência Respiratória
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA