insight.jci.org Open in urlscan Pro
96.126.107.125  Public Scan

Submitted URL: http://insight.jci.org/
Effective URL: https://insight.jci.org/
Submission: On March 22 via api from US — Scanned from DE

Form analysis 4 forms found in the DOM

GET /search/results

<form action="/search/results" accept-charset="UTF-8" method="get">
  <input type="text" name="q" id="q" value="" placeholder="Search JCI Insight">
  <input type="image" src="/assets/search-black-ba9b554d6f74b1c93d6e6ab71d1c9830c18a20fc6b7e72393f136f5d875141ac.png" value="">
</form>

GET /search/results

<form action="/search/results" accept-charset="UTF-8" method="get">
  <div class="row collapse" id="search-div-offcanvas">
    <div class="small-8 columns">
      <input name="q" placeholder="Search the JCI Insight" type="text">
    </div>
    <div class="small-4 columns">
      <input type="image" src="/assets/common/search-white-530f3f95b9080d73eba51eaeffdf1a3922af42ccc277a2d1d987b8aa24423c96.png" id="search-icon-offcanvas">
    </div>
  </div>
</form>

GET https://notices.jci.org/subscribers/new

<form action="https://notices.jci.org/subscribers/new" method="get">
  <input name="utm_source" type="hidden" value="jci">
  <input name="utm_medium" type="hidden" value="web">
  <input name="utm_campaign" type="hidden" value="email_signup">
  <input name="utm_content" type="hidden" value="homepage">
  <div class="row">
    <div class="small-12 medium-9 columns">
      <input name="email_address" placeholder="Your email address" required="" type="text">
    </div>
    <div class="small-12 medium-3 columns">
      <input class="button tiny orange" type="submit" value="Sign up">
    </div>
  </div>
</form>

GET https://notices.jci.org/subscribers/new

<form action="https://notices.jci.org/subscribers/new" method="get">
  <input name="utm_source" type="hidden" value="insight">
  <input name="utm_medium" type="hidden" value="web">
  <input name="utm_campaign" type="hidden" value="email_signup">
  <input name="utm_content" type="hidden" value="footer">
  <div class="row">
    <div class="small-12 medium-9 columns">
      <input name="email_address" placeholder="Your email address" required="" type="text">
    </div>
    <div class="small-12 medium-3 columns">
      <input class="button tiny orange" type="submit" value="Sign up">
    </div>
  </div>
</form>

Text Content

Go to The Journal of Clinical Investigation
 * About
 * Editors
 * Consulting Editors
 * For authors
 * Publication ethics
 * Publication alerts by email
 * Transfers
 * Advertising
 * Job board
 * Contact

 * Current issue
 * Past issues
 * By specialty
   
   * BACK
   
   * COVID-19
   * Cardiology
   * Immunology
   * Metabolism
   * Nephrology
   * Oncology
   * Pulmonology
   * All ...
 * Videos
 * Collections
   
   * BACK
   
   * 
   * Resource and Technical Advances
   * Clinical Medicine
   * Reviews
   * Editorials
   * Perspectives
   * JCI This Month
   * Top read articles

 * 




 * JCI Insight
 * 
 * Current issue
 * Past issues
 * Specialties
 * Collections
 * In-Press Preview
 * 
 * 
 * 
 * 
 * Editorials
 * Viewpoint
 * JCI This Month
 * Top read articles
 * Journal Details
 * About
 * Editors
 * Consulting Editors
 * For authors
 * Publication ethics
 * Publication alerts by email
 * Transfers
 * Advertising
 * Job board
 * Contact

ISSUE PUBLISHED MARCH 8, 2024

 * Volume 9, Issue 5
 * Previous Issue

GO TO SECTION:

 * Research Articles
 * Technical Advances
 * Physician-Scientist Development
 * Corrigendum
 * Editorial

ENDOTHELIAL TDP-43 CONTROLS SPROUTING ANGIOGENESIS AND VASCULAR BARRIER
INTEGRITY, AND ITS DELETION TRIGGERS NEUROINFLAMMATION

Arribas et al. report roles for TDP-43 in the growth and stabilization of CNS
vessels and identify endothelial TDP-43 as a factor potentially contributing to
the vascular disorders and inflammation observed in patients diagnosed with
TDP-43–associated diseases. The cover image shows blood–brain barrier disruption
and neuroinflammation in a murine TDP-43iΔEC brain section stained for IB4
(green), the microglial marker Iba1 (light blue), and Ter119 (red blood cell
marker; red).

Technical Advances

MODELING SKELETAL DYSPLASIA IN HURLER SYNDROME USING PATIENT-DERIVED BONE MARROW
OSTEOPROGENITOR CELLS

Samantha Donsante, … , Marta Serafini, Mara Riminucci
Samantha Donsante, … , Marta Serafini, Mara Riminucci
Published March 8, 2024
Citation Information: JCI Insight. 2024;9(5):e173449.
https://doi.org/10.1172/jci.insight.173449.
View: Text | PDF



MODELING SKELETAL DYSPLASIA IN HURLER SYNDROME USING PATIENT-DERIVED BONE MARROW
OSTEOPROGENITOR CELLS

 * Text
 * PDF

ABSTRACT

Dysostosis multiplex is a major cause of morbidity in Hurler syndrome
(mucopolysaccharidosis type IH [MPS IH], OMIM #607014) because currently
available therapies have limited success in its prevention and reversion.
Unfortunately, the elucidation of skeletal pathogenesis in MPS IH is limited by
difficulties in obtaining bone specimens from pediatric patients and poor
reproducibility in animal models. Thus, the application of experimental systems
that can be used to dissect cellular and molecular mechanisms underlying the
skeletal phenotype of MPS IH patients and to identify effective therapies is
highly needed. Here, we adopted in vitro/in vivo systems based on
patient-derived bone marrow stromal cells to generate cartilaginous pellets and
bone rudiments. Interestingly, we observed that heparan sulphate accumulation
compromised the remodeling of MPS IH cartilage into other skeletal tissues and
other critical aspects of the endochondral ossification process. We also noticed
that MPS IH hypertrophic cartilage was characterized by dysregulation of
signaling pathways controlling cartilage hypertrophy and fate, extracellular
matrix organization, and glycosaminoglycan metabolism. Our study demonstrates
that the cartilaginous pellet–based system is a valuable tool to study MPS IH
dysostosis and to develop new therapeutic approaches for this hard-to-treat
aspect of the disease. Finally, our approach may be applied for modeling other
genetic skeletal disorders.

AUTHORS

Samantha Donsante, Alice Pievani, Biagio Palmisano, Melissa Finamore, Grazia
Fazio, Alessandro Corsi, Andrea Biondi, Shunji Tomatsu, Rocco Piazza, Marta
Serafini, Mara Riminucci

×

--------------------------------------------------------------------------------

ADAMTSL2 MUTATIONS DETERMINE THE PHENOTYPIC SEVERITY IN GELEOPHYSIC DYSPLASIA

Vladimir Camarena, … , Katherina Walz, Mustafa Tekin
Vladimir Camarena, … , Katherina Walz, Mustafa Tekin
Published February 1, 2024
Citation Information: JCI Insight. 2024;9(5):e174417.
https://doi.org/10.1172/jci.insight.174417.
View: Text | PDF



ADAMTSL2 MUTATIONS DETERMINE THE PHENOTYPIC SEVERITY IN GELEOPHYSIC DYSPLASIA

 * Text
 * PDF

ABSTRACT

Geleophysic dysplasia-1 (GD1) is an autosomal recessive disorder caused by
ADAMTS-like 2 (ADAMTSL2) variants. It is characterized by distinctive facial
features, limited joint mobility, short stature, brachydactyly, and
life-threatening cardiorespiratory complications. The clinical spectrum spans
from perinatal lethality to milder adult phenotypes. We developed and
characterized cellular and mouse models, to replicate the genetic profile of a
patient who is compound heterozygous for 2 ADAMTSL2 variants, namely p.R61H and
p.A165T. The impairment of ADAMTSL2 secretion was observed in both variants, but
p.A165T exhibited a more severe impact. Mice carrying different allelic
combinations revealed a spectrum of phenotypic severity, from lethality in
knockout homozygotes to mild growth impairment observed in adult p.R61H
homozygotes. Homozygous and hemizygous p.A165T mice survived but displayed
severe respiratory and cardiac dysfunction. The respiratory dysfunction mainly
affected the expiration phase, and some of these animals had microscopic
post-obstructive pneumonia. Echocardiograms and MRI studies revealed a
significant systolic dysfunction, accompanied by a reduction of the aortic root
size. Histology verified the presence of hypertrophic cardiomyopathy with
myocyte hypertrophy, chondroid metaplasia, and mild interstitial fibrosis. This
study revealed a substantial correlation between the degree of impaired ADAMTSL2
secretion and the severity of the observed phenotype in GD1.

AUTHORS

Vladimir Camarena, Monique M. Williams, Alejo A. Morales, Mohammad F. Zafeer,
Okan V. Kilic, Ali Kamiar, Clemer Abad, Monica A. Rasmussen, Laurence M. Briski,
LéShon Peart, Guney Bademci, Deborah S. Barbouth, Sarah Smithson, Gaofeng Wang,
Lina A. Shehadeh, Katherina Walz, Mustafa Tekin

×

--------------------------------------------------------------------------------

A MOUSE MODEL OF ZHU-TOKITA-TAKENOUCHI-KIM SYNDROME REVEALS INDISPENSABLE SON
FUNCTIONS IN ORGAN DEVELOPMENT AND HEMATOPOIESIS

Lana Vukadin, … , Ssang-Taek Steve Lim, Eun-Young Erin Ahn
Lana Vukadin, … , Ssang-Taek Steve Lim, Eun-Young Erin Ahn
Published January 30, 2024
Citation Information: JCI Insight. 2024;9(5):e175053.
https://doi.org/10.1172/jci.insight.175053.
View: Text | PDF



A MOUSE MODEL OF ZHU-TOKITA-TAKENOUCHI-KIM SYNDROME REVEALS INDISPENSABLE SON
FUNCTIONS IN ORGAN DEVELOPMENT AND HEMATOPOIESIS

 * Text
 * PDF

ABSTRACT

Rare diseases are underrepresented in biomedical research, leading to
insufficient awareness. Zhu-Tokita-Takenouchi-Kim (ZTTK) syndrome is a rare
disease caused by genetic alterations that result in heterozygous loss of
function of SON. While patients with ZTTK syndrome live with numerous symptoms,
the lack of model organisms hampers our understanding of SON and this complex
syndrome. Here, we developed Son haploinsufficiency (Son+/–) mice as a model of
ZTTK syndrome and identified the indispensable roles of Son in organ development
and hematopoiesis. Son+/– mice recapitulated clinical symptoms of ZTTK syndrome,
including growth retardation, cognitive impairment, skeletal abnormalities, and
kidney agenesis. Furthermore, we identified hematopoietic abnormalities in
Son+/– mice, including leukopenia and immunoglobulin deficiency, similar to
those observed in human patients. Surface marker analyses and single-cell
transcriptome profiling of hematopoietic stem and progenitor cells revealed that
Son haploinsufficiency shifted cell fate more toward the myeloid lineage but
compromised lymphoid lineage development by reducing genes required for lymphoid
and B cell lineage specification. Additionally, Son haploinsufficiency caused
inappropriate activation of erythroid genes and impaired erythropoiesis. These
findings highlight the importance of the full gene expression of Son in multiple
organs. Our model serves as an invaluable research tool for this rare disease
and related disorders associated with SON dysfunction.

AUTHORS

Lana Vukadin, Bohye Park, Mostafa Mohamed, Huashi Li, Amr Elkholy, Alex
Torrelli-Diljohn, Jung-Hyun Kim, Kyuho Jeong, James M. Murphy, Caitlin A.
Harvey, Sophia Dunlap, Leah Gehrs, Hanna Lee, Hyung-Gyoon Kim, Jay Prakash Sah,
Seth N. Lee, Denise Stanford, Robert A. Barrington, Jeremy B. Foote, Anna G.
Sorace, Robert S. Welner, Blake E. Hildreth III, Ssang-Taek Steve Lim, Eun-Young
Erin Ahn

×
Physician-Scientist Development

INTERVENTIONS TO SUPPORT FELLOWSHIP APPLICATION SUCCESS AMONG PREDOCTORAL
PHYSICIAN-SCIENTISTS

Reiko Maki Fitzsimonds, … , Fred S. Gorelick, Barbara I. Kazmierczak
Reiko Maki Fitzsimonds, … , Fred S. Gorelick, Barbara I. Kazmierczak
Published March 8, 2024
Citation Information: JCI Insight. 2024;9(5):e175857.
https://doi.org/10.1172/jci.insight.175857.
View: Text | PDF



INTERVENTIONS TO SUPPORT FELLOWSHIP APPLICATION SUCCESS AMONG PREDOCTORAL
PHYSICIAN-SCIENTISTS

 * Text
 * PDF

ABSTRACT

A critical element of physician-scientist training is the development and
practice of core competencies that promote success in research careers. The
ability to develop compelling training and research proposals is one such
foundational skill. The NIH Ruth L. Kirschstein National Research Service Award
(NRSA) individual fellowship for dual-degree students (F30, F31, or
F31-Diversity) creates an ideal opportunity to provide formal instruction in
grant-writing skills to physician-scientists early in training. In the guided
process of preparing a predoctoral fellowship application, students learn to
formulate clear short- and long-term research and training goals; construct a
comprehensive, well-reasoned, and rigorous proposal; become familiar with
funding agency priorities; and gain strategic insights into the peer review
system. Beyond building scientific writing skills, the application process for
an NRSA F30 or F31 is an opportunity for trainees to strengthen mentor-mentee
relationships, identify learning opportunities key to their scientific
development, and build effective research and mentoring teams. These skills also
apply to developing future postdoctoral mentored K applications or faculty
research program grants. Here, we outline key features of the structured
proposal development training developed for students in the Yale MD-PhD Program
and review outcomes associated with its implementation.

AUTHORS

Reiko Maki Fitzsimonds, Fred S. Gorelick, Barbara I. Kazmierczak

×
Editorial

PHYSICIAN-SCIENTIST DEVELOPMENT: A NEW CATEGORY FOR SUPPORTING ALL STAGES OF THE
PHYSICIAN-SCIENTIST PIPELINE

Kathleen L. Collins
Kathleen L. Collins
Published March 8, 2024
Citation Information: JCI Insight. 2024;9(5):e179940.
https://doi.org/10.1172/jci.insight.179940.
View: Text | PDF


PHYSICIAN-SCIENTIST DEVELOPMENT: A NEW CATEGORY FOR SUPPORTING ALL STAGES OF THE
PHYSICIAN-SCIENTIST PIPELINE

 * Text
 * PDF

ABSTRACT



AUTHORS

Kathleen L. Collins

×
Research Articles

A T CELL–BASED SARS-COV-2 SPIKE PROTEIN VACCINE PROVIDES PROTECTION WITHOUT
ANTIBODIES

Juan Shi, … , Liang Qiao, Lanying Du
Juan Shi, … , Liang Qiao, Lanying Du
Published January 23, 2024
Citation Information: JCI Insight. 2024;9(5):e155789.
https://doi.org/10.1172/jci.insight.155789.
View: Text | PDF



A T CELL–BASED SARS-COV-2 SPIKE PROTEIN VACCINE PROVIDES PROTECTION WITHOUT
ANTIBODIES

 * Text
 * PDF

ABSTRACT

SARS-CoV-2 spike–based vaccines are used to control the COVID-19 pandemic.
However, emerging variants have become resistant to antibody neutralization and
further mutations may lead to full resistance. We tested whether T cells alone
could provide protection without antibodies. We designed a T cell–based vaccine
in which SARS-CoV-2 spike sequences were rearranged and attached to ubiquitin.
Immunization of mice with the vaccine induced no specific antibodies, but strong
specific T cell responses. We challenged mice with SARS-CoV-2 wild-type strain
or an Omicron variant after the immunization and monitored survival or viral
titers in the lungs. The mice were significantly protected against death and
weight loss caused by the SARS-CoV-2 wild-type strain, and the viral titers in
the lungs of mice challenged with the SARS-CoV-2 wild-type strain or the Omicron
variant were significantly reduced. Importantly, depletion of CD4+ or CD8+ T
cells led to significant loss of the protection. Our analyses of spike protein
sequences of the variants indicated that fewer than one-third presented by
dominant HLA alleles were mutated and that most of the mutated epitopes were in
the subunit 1 region. As the subunit 2 region is conservative, the vaccines
targeting spike protein are expected to protect against future variants due to
the T cell responses.

AUTHORS

Juan Shi, Jian Zheng, Xiujuan Zhang, Wanbo Tai, Ryan Compas, Jack Deno, Natalie
Jachym, Abhishek K. Verma, Gang Wang, Xiaoqing Guan, Abby E. Odle, Yushun Wan,
Fang Li, Stanley Perlman, Liang Qiao, Lanying Du

×

--------------------------------------------------------------------------------

HUMAN MAIT CELLS INHIBIT ALLOREACTIVE T CELL RESPONSES AND PROTECT AGAINST ACUTE
GRAFT-VERSUS-HOST DISEASE

Nana Talvard-Balland, … , Olivier Lantz, Sophie Caillat-Zucman
Nana Talvard-Balland, … , Olivier Lantz, Sophie Caillat-Zucman
Published February 1, 2024
Citation Information: JCI Insight. 2024;9(5):e166310.
https://doi.org/10.1172/jci.insight.166310.
View: Text | PDF



HUMAN MAIT CELLS INHIBIT ALLOREACTIVE T CELL RESPONSES AND PROTECT AGAINST ACUTE
GRAFT-VERSUS-HOST DISEASE

 * Text
 * PDF

ABSTRACT

Adoptive transfer of immunoregulatory cells can prevent or ameliorate
graft-versus-host disease (GVHD), which remains the main cause of nonrelapse
mortality after allogeneic hematopoietic stem cell transplantation.
Mucosal-associated invariant T (MAIT) cells were recently associated with tissue
repair capacities and with lower rates of GVHD in humans. Here, we analyzed the
immunosuppressive effect of MAIT cells in an in vitro model of alloreactivity
and explored their adoptive transfer in a preclinical xenogeneic GVHD model. We
found that MAIT cells, whether freshly purified or short-term expanded,
dose-dependently inhibited proliferation and activation of alloreactive T cells.
In immunodeficient mice injected with human PBMCs, MAIT cells greatly delayed
GVHD onset and decreased severity when transferred early after PBMC injection
but could also control ongoing GVHD when transferred at delayed time points.
This effect was associated with decreased proliferation and effector function of
human T cells infiltrating tissues of diseased mice and was correlated with
lower circulating IFN-γ and TNF-α levels and increased IL-10 levels. MAIT cells
acted partly in a contact-dependent manner, which likely required direct
interaction of their T cell receptor with MHC class I–related molecule (MR1)
induced on host-reactive T cells. These results support the setup of clinical
trials using MAIT cells as universal therapeutic tools to control severe GVHD or
mucosal inflammatory disorders.

AUTHORS

Nana Talvard-Balland, Marion Lambert, Mathieu F. Chevalier, Norbert Minet,
Marion Salou, Marie Tourret, Armelle Bohineust, Idan Milo, Véronique Parietti,
Thomas Yvorra, Gérard Socié, Olivier Lantz, Sophie Caillat-Zucman

×

--------------------------------------------------------------------------------

AGE-RELATED DYSREGULATION OF INTESTINAL EPITHELIUM FUCOSYLATION IS LINKED TO AN
INCREASED RISK OF COLON CANCER

Zhihan Wang, … , Donald A. Jurivich, Ramkumar Mathur
Zhihan Wang, … , Donald A. Jurivich, Ramkumar Mathur
Published January 30, 2024
Citation Information: JCI Insight. 2024;9(5):e167676.
https://doi.org/10.1172/jci.insight.167676.
View: Text | PDF



AGE-RELATED DYSREGULATION OF INTESTINAL EPITHELIUM FUCOSYLATION IS LINKED TO AN
INCREASED RISK OF COLON CANCER

 * Text
 * PDF

ABSTRACT

Colon cancer affects people of all ages. However, its frequency, as well as the
related morbidity and mortality, are high among older adults. The complex
physiological changes in the aging gut substantially limit the development of
cancer therapies. Here, we identify a potentially unique intestinal
microenvironment that is linked with an increased risk of colon cancer in older
adults. Our findings show that aging markedly influenced persistent fucosylation
of the apical surfaces of intestinal epithelial cells, which resulted in a
favorable environment for tumor growth. Furthermore, our findings shed light on
the importance of the host-commensal interaction, which facilitates the
dysregulation of fucosylation and promotes tumor growth as people get older. We
analyzed colonic microbial populations at the species level to find changes
associated with aging that could contribute to the development of colon cancer.
Analysis of single-cell RNA-sequencing data from previous publications
identified distinct epithelial cell subtypes involved in dysregulated
fucosylation in older adults. Overall, our study provides compelling evidence
that excessive fucosylation is associated with the development of colon cancer,
that age-related changes increase vulnerability to colon cancer, and that a
dysbiosis in microbial diversity and metabolic changes in the homeostasis of
older mice dysregulate fucosylation levels with age.

AUTHORS

Zhihan Wang, Pan Gao, Kai Guo, Grace Schirrick, Jappreet Singh Gill, Jett Weis,
Abby Lund Da Costa, Mansib Rahman, Het Mehta, Julia Fleecs, Shilpi Jain, Trishna
Debnath, Junguk Hur, Nadeem Khan, Robert Sticca, Holly M. Brown-Borg, Donald A.
Jurivich, Ramkumar Mathur

×

--------------------------------------------------------------------------------

NOREPINEPHRINE INDUCES ANOIKIS RESISTANCE IN HIGH-GRADE SEROUS OVARIAN CANCER
PRECURSOR CELLS

Hunter D. Reavis, … , Laura M. Sanchez, Ronny Drapkin
Hunter D. Reavis, … , Laura M. Sanchez, Ronny Drapkin
Published January 25, 2024
Citation Information: JCI Insight. 2024;9(5):e170961.
https://doi.org/10.1172/jci.insight.170961.
View: Text | PDF



NOREPINEPHRINE INDUCES ANOIKIS RESISTANCE IN HIGH-GRADE SEROUS OVARIAN CANCER
PRECURSOR CELLS

 * Text
 * PDF

ABSTRACT

High-grade serous carcinoma (HGSC) is the most lethal gynecological malignancy
in the United States. Late diagnosis and the emergence of chemoresistance have
prompted studies into how the tumor microenvironment, and more recently tumor
innervation, may be leveraged for HGSC prevention and interception. In addition
to stess-induced sources, concentrations of the sympathetic neurotransmitter
norepinephrine (NE) in the ovary increase during ovulation and after menopause.
Importantly, NE exacerbates advanced HGSC progression. However, little is known
about the role of NE in early disease pathogenesis. Here, we investigated the
role of NE in instigating anchorage independence and micrometastasis of
preneoplastic lesions from the fallopian tube epithelium (FTE) to the ovary, an
essential step in HGSC onset. We found that in the presence of NE, FTE cell
lines were able to survive in ultra-low-attachment (ULA) culture in a
β-adrenergic receptor–dependent (β-AR–dependent) manner. Importantly, spheroid
formation and cell viability conferred by treatment with physiological sources
of NE were abrogated using the β-AR blocker propranolol. We have also identified
that NE-mediated anoikis resistance may be attributable to downregulation of
colony-stimulating factor 2. These findings provide mechanistic insight and
identify targets that may be regulated by ovary-derived NE in early HGSC.

AUTHORS

Hunter D. Reavis, Stefan M. Gysler, Grace B. McKenney, Matthew Knarr, Hannah J.
Lusk, Priyanka Rawat, Hannah S. Rendulich, Marilyn A. Mitchell, Dara S. Berger,
Jamie S. Moon, Suyeon Ryu, Monica Mainigi, Marcin P. Iwanicki, Dave S. Hoon,
Laura M. Sanchez, Ronny Drapkin

×

--------------------------------------------------------------------------------

DDIT4L REGULATES MITOCHONDRIAL AND INNATE IMMUNE ACTIVITIES IN EARLY LIFE

Christina Michalski, … , Ramon I. Klein Geltink, Pascal M. Lavoie
Christina Michalski, … , Ramon I. Klein Geltink, Pascal M. Lavoie
Published February 6, 2024
Citation Information: JCI Insight. 2024;9(5):e172312.
https://doi.org/10.1172/jci.insight.172312.
View: Text | PDF



DDIT4L REGULATES MITOCHONDRIAL AND INNATE IMMUNE ACTIVITIES IN EARLY LIFE

 * Text
 * PDF

ABSTRACT

Pattern recognition receptor responses are profoundly attenuated before the
third trimester of gestation in the relatively low-oxygen human fetal
environment. However, the mechanisms regulating these responses are
uncharacterized. Herein, genome-wide transcription and functional metabolic
experiments in primary neonatal monocytes linked the negative mTOR regulator
DDIT4L to metabolic stress, cellular bioenergetics, and innate immune activity.
Using genetically engineered monocytic U937 cells, we confirmed that DDIT4L
overexpression altered mitochondrial dynamics, suppressing their activity, and
blunted LPS-induced cytokine responses. We also showed that monocyte
mitochondrial function is more restrictive in earlier gestation, resembling the
phenotype of DDIT4L-overexpressing U937 cells. Gene expression analyses in
neonatal granulocytes and lung macrophages in preterm infants confirmed
upregulation of the DDIT4L gene in the early postnatal period and also suggested
a potential protective role against inflammation-associated chronic neonatal
lung disease. Taken together, these data show that DDIT4L regulates
mitochondrial activity and provide what we believe to be the first direct
evidence for its potential role supressing innate immune activity in myeloid
cells during development.

AUTHORS

Christina Michalski, Claire Cheung, Ju Hee Oh, Emma Ackermann, Constantin R.
Popescu, Anne-Sophie Archambault, Martin A. Prusinkiewicz, Rachel Da Silva,
Abdelilah Majdoubi, Marina Viñeta Paramo, Rui Yang Xu, Frederic Reicherz,
Annette E. Patterson, Liam Golding, Ashish A. Sharma, Chinten J. Lim, Paul C.
Orban, Ramon I. Klein Geltink, Pascal M. Lavoie

×

--------------------------------------------------------------------------------

PREX1 IMPROVES HOMEOSTATIC PROLIFERATION TO MAINTAIN A NAIVE CD4+ T CELL
COMPARTMENT IN OLDER AGE

Huimin Zhang, … , Cornelia M. Weyand, Jӧrg J. Goronzy
Huimin Zhang, … , Cornelia M. Weyand, Jӧrg J. Goronzy
Published February 8, 2024
Citation Information: JCI Insight. 2024;9(5):e172848.
https://doi.org/10.1172/jci.insight.172848.
View: Text | PDF



PREX1 IMPROVES HOMEOSTATIC PROLIFERATION TO MAINTAIN A NAIVE CD4+ T CELL
COMPARTMENT IN OLDER AGE

 * Text
 * PDF

ABSTRACT

The human adult immune system maintains normal T cell counts and compensates for
T cell loss throughout life, mainly through peripheral homeostatic proliferation
after the ability of the thymus to generate new T cells has rapidly declined at
adolescence. This process is mainly driven by STAT5-activating cytokines, most
importantly IL-7, and is very effective in maintaining a large naive CD4+ T cell
compartment into older age. Here, we describe that naive CD4+ T cells undergo
adaptations to optimize IL-7 responses by upregulating the guanine-nucleotide
exchange factor PREX1 in older age. PREX1 promotes nuclear translocation of
phosphorylated STAT5, thereby supporting homeostatic proliferation in response
to IL-7. Through the same mechanism, increased expression of PREX1 also biases
naive cells to differentiate into effector T cells. These findings are
consistent with the concept that primarily beneficial adaptations during aging,
i.e., improved homeostasis, account for unfavorable functions of the aged immune
system, in this case biased differentiation.

AUTHORS

Huimin Zhang, Hirohisa Okuyama, Abhinav Jain, Rohit R. Jadhav, Bowen Wu, Ines
Sturmlechner, Jose Morales, Shozo Ohtsuki, Cornelia M. Weyand, Jӧrg J. Goronzy

×

--------------------------------------------------------------------------------

TREGS FROM HUMAN BLOOD DIFFERENTIATE INTO NONLYMPHOID TISSUE–RESIDENT EFFECTOR
CELLS UPON TNFR2 COSTIMULATION

Mark Mensink, … , Sander de Kivit, Jannie Borst
Mark Mensink, … , Sander de Kivit, Jannie Borst
Published January 30, 2024
Citation Information: JCI Insight. 2024;9(5):e172942.
https://doi.org/10.1172/jci.insight.172942.
View: Text | PDF



TREGS FROM HUMAN BLOOD DIFFERENTIATE INTO NONLYMPHOID TISSUE–RESIDENT EFFECTOR
CELLS UPON TNFR2 COSTIMULATION

 * Text
 * PDF

ABSTRACT

Tregs can facilitate transplant tolerance and attenuate autoimmune and
inflammatory diseases. Therefore, it is clinically relevant to stimulate Treg
expansion and function in vivo and to create therapeutic Treg products in vitro.
We report that TNF receptor 2 (TNFR2) is a unique costimulus for naive,
thymus-derived Tregs (tTregs) from human blood that promotes their
differentiation into nonlymphoid tissue–resident (NLT-resident) effector Tregs,
without Th-like polarization. In contrast, CD28 costimulation maintains a
lymphoid tissue–resident (LT-resident) Treg phenotype. We base this conclusion
on transcriptome and proteome analysis of TNFR2- and CD28-costimulated CD4+
tTregs and conventional T cells (Tconvs), followed by bioinformatic comparison
with published transcriptomic Treg signatures from NLT and LT in health and
disease, including autoimmunity and cancer. These analyses illuminate that TNFR2
costimulation promoted tTreg capacity for survival, migration,
immunosuppression, and tissue regeneration. Functional studies confirmed
improved migratory ability of TNFR2-costimulated tTregs. Flow cytometry
validated the presence of the TNFR2-driven tTreg signature in effector/memory
Tregs from the human placenta, as opposed to blood. Thus, TNFR2 can be exploited
as a driver of NLT-resident tTreg differentiation for adoptive cell therapy or
antibody-based immunomodulation in human disease.

AUTHORS

Mark Mensink, Lotte J. Verleng, Ellen Schrama, George M.C. Janssen, Rayman T.N.
Tjokrodirijo, Peter A. van Veelen, Qinyue Jiang, M. Fernanda Pascutti,
Marie-Louise van der Hoorn, Michael Eikmans, Sander de Kivit, Jannie Borst

×

--------------------------------------------------------------------------------

MACROPHAGE-ENRICHED SECTM1A PROMOTES EFFICIENT EFFEROCYTOSIS TO ATTENUATE
ISCHEMIA/REPERFUSION-INDUCED CARDIAC INJURY

Xiaohong Wang, … , Wei Huang, Guo-Chang Fan
Xiaohong Wang, … , Wei Huang, Guo-Chang Fan
Published March 8, 2024
Citation Information: JCI Insight. 2024;9(5):e173832.
https://doi.org/10.1172/jci.insight.173832.
View: Text | PDF



MACROPHAGE-ENRICHED SECTM1A PROMOTES EFFICIENT EFFEROCYTOSIS TO ATTENUATE
ISCHEMIA/REPERFUSION-INDUCED CARDIAC INJURY

 * Text
 * PDF

ABSTRACT

Efficient clearance and degradation of apoptotic cardiomyocytes by macrophages
(collectively termed efferocytosis) is critical for inflammation resolution and
restoration of cardiac function after myocardial ischemia/reperfusion (I/R).
Here, we define secreted and transmembrane protein 1a (Sectm1a), a cardiac
macrophage–enriched gene, as a modulator of macrophage efferocytosis in
I/R-injured hearts. Upon myocardial I/R, Sectm1a-KO mice exhibited impaired
macrophage efferocytosis, leading to massive accumulation of apoptotic
cardiomyocytes, cardiac inflammation, fibrosis, and consequently, exaggerated
cardiac dysfunction. By contrast, therapeutic administration of recombinant
SECTM1A protein significantly enhanced macrophage efferocytosis and improved
cardiac function. Mechanistically, SECTM1A could elicit autocrine effects on the
activation of glucocorticoid-induced TNF receptor (GITR) at the surface of
macrophages, leading to the upregulation of liver X receptor α (LXRα) and its
downstream efferocytosis-related genes and lysosomal enzyme genes. Our study
suggests that Sectm1a-mediated activation of the Gitr/LXRα axis could be a
promising approach to enhance macrophage efferocytosis for the treatment of
myocardial I/R injury.

AUTHORS

Xiaohong Wang, Wa Du, Yutian Li, Hui-Hui Yang, Yu Zhang, Rubab Akbar, Hannah
Morgan, Tianqing Peng, Jing Chen, Sakthivel Sadayappan, Yueh-Chiang Hu, Yanbo
Fan, Wei Huang, Guo-Chang Fan

×

--------------------------------------------------------------------------------

LOSS OF T FOLLICULAR REGULATORY CELL–DERIVED IL-1R2 AUGMENTS GERMINAL CENTER
REACTIONS VIA INCREASED IL-1

Katerina Pyrillou, … , Ziad Mallat, Murray C.H. Clarke
Katerina Pyrillou, … , Ziad Mallat, Murray C.H. Clarke
Published February 8, 2024
Citation Information: JCI Insight. 2024;9(5):e174005.
https://doi.org/10.1172/jci.insight.174005.
View: Text | PDF



LOSS OF T FOLLICULAR REGULATORY CELL–DERIVED IL-1R2 AUGMENTS GERMINAL CENTER
REACTIONS VIA INCREASED IL-1

 * Text
 * PDF

ABSTRACT

Inappropriate immune activity is key in the pathogenesis of multiple diseases,
and it is typically driven by excess inflammation and/or autoimmunity. IL-1 is
often the effector owing to its powerful role in both innate and adaptive
immunity, and, thus, it is tightly controlled at multiple levels. IL-1R2
antagonizes IL-1, but effects of losing this regulation are unknown. We found
that IL-1R2 resolves inflammation by rapidly scavenging free IL-1. Specific
IL-1R2 loss in germinal center (GC) T follicular regulatory (Tfr) cells
increased the GC response after a first, but not booster, immunization, with an
increase in T follicular helper (Tfh) cells, GC B cells, and antigen-specific
antibodies, which was reversed upon IL-1 blockade. However, IL-1 signaling is
not obligate for GC reactions, as WT and Il1r1–/– mice showed equivalent
phenotypes, suggesting that GC IL-1 is normally restrained by IL-1R2.
Fascinatingly, germline Il1r2–/– mice did not show this phenotype, but
conditional Il1r2 deletion in adulthood recapitulated it, implying that
compensation during development counteracts IL-1R2 loss. Finally, patients with
ulcerative colitis or Crohn’s disease had lower serum IL-1R2. All together, we
show that IL-1R2 controls important aspects of innate and adaptive immunity and
that IL-1R2 level may contribute to human disease propensity and/or progression.

AUTHORS

Katerina Pyrillou, Melanie Humphry, Lauren A. Kitt, Amanda Rodgers, Meritxell
Nus, Martin R. Bennett, Kenneth G.C. Smith, Paul A. Lyons, Ziad Mallat, Murray
C.H. Clarke

×

--------------------------------------------------------------------------------

HIGH-MOBILITY GROUP BOX 1 INCREASES PLATELET SURFACE P2Y12 AND PLATELET
ACTIVATION IN SICKLE CELL DISEASE

Deirdre Nolfi-Donegan, … , Cheryl A. Hillery, Sruti Shiva
Deirdre Nolfi-Donegan, … , Cheryl A. Hillery, Sruti Shiva
Published March 8, 2024
Citation Information: JCI Insight. 2024;9(5):e174575.
https://doi.org/10.1172/jci.insight.174575.
View: Text | PDF



HIGH-MOBILITY GROUP BOX 1 INCREASES PLATELET SURFACE P2Y12 AND PLATELET
ACTIVATION IN SICKLE CELL DISEASE

 * Text
 * PDF

ABSTRACT

Thrombosis and inflammation are intimately linked and synergistically contribute
to the pathogenesis of numerous thromboinflammatory diseases, including sickle
cell disease (SCD). While platelets are central to thrombogenesis and
inflammation, the molecular mechanisms of crosstalk between the 2 remain
elusive. High-mobility group box 1 (HMGB1) regulates inflammation and stimulates
platelet activation through Toll-like receptor 4. However, it remains unclear
whether HMGB1 modulates other thrombotic agonists to regulate platelet
activation. Herein, using human platelets, we demonstrate that HMGB1
significantly enhanced ADP-mediated platelet activation. Furthermore, inhibition
of the purinergic receptor P2Y12 attenuated HMGB1-dependent platelet activation.
Mechanistically, we show that HMGB1 stimulated ADP secretion, while
concomitantly increasing P2Y12 levels at the platelet membrane. We show that in
SCD patients, increased plasma HMGB1 levels were associated with heightened
platelet activation and surface P2Y12 expression. Treatment of healthy platelets
with plasma from SCD patients enhanced platelet activation and surface P2Y12,
and increased sensitivity to ADP-mediated activation, and these effects were
linked to plasma HMGB1. We conclude that HMGB1-mediated platelet activation
involves ADP-dependent P2Y12 signaling, and HMGB1 primes platelets for ADP
signaling. This complementary agonism between ADP and HMGB1 furthers the
understanding of thromboinflammatory signaling in conditions such as SCD, and
provides insight for therapeutic P2Y12 inhibition.

AUTHORS

Deirdre Nolfi-Donegan, Gowtham K. Annarapu, Claudette St. Croix, Michael
Calderon, Cheryl A. Hillery, Sruti Shiva

×

--------------------------------------------------------------------------------

ACTIVATOR PROTEIN TRANSCRIPTION FACTORS COORDINATE HUMAN IL-33 EXPRESSION FROM
NONCANONICAL PROMOTERS IN CHRONIC AIRWAY DISEASE

Heather E. Raphael, … , Bo Zhang, Jen Alexander-Brett
Heather E. Raphael, … , Bo Zhang, Jen Alexander-Brett
Published March 8, 2024
Citation Information: JCI Insight. 2024;9(5):e174786.
https://doi.org/10.1172/jci.insight.174786.
View: Text | PDF



ACTIVATOR PROTEIN TRANSCRIPTION FACTORS COORDINATE HUMAN IL-33 EXPRESSION FROM
NONCANONICAL PROMOTERS IN CHRONIC AIRWAY DISEASE

 * Text
 * PDF

ABSTRACT

IL-33 is a cytokine central to type 2 immune pathology in chronic airway
disease. This cytokine is abundantly expressed in the respiratory epithelium and
increased in disease, but how expression is regulated is undefined. Here we show
that increased IL33 expression occurs from multiple noncanonical promoters in
human chronic obstructive pulmonary disease (COPD), and it facilitates
production of alternatively spliced isoforms in airway cells. We found that
phorbol 12-myristate 13-acetate (PMA) can activate IL33 promoters through
protein kinase C in primary airway cells and lines. Transcription factor (TF)
binding arrays combined with RNA interference identified activator protein (AP)
TFs as regulators of baseline and induced IL33 promoter activity. ATAC-Seq and
ChIP-PCR identified chromatin accessibility and differential TF binding as
additional control points for transcription from noncanonical promoters. In
support of a role for these TFs in COPD pathogenesis, we found that AP-2
(TFAP2A, TFAP2C) and AP-1 (FOS and JUN) family members are upregulated in human
COPD specimens. This study implicates integrative and pioneer TFs in regulating
IL33 promoters and alternative splicing in human airway basal cells. Our work
reveals a potentially novel approach for targeting IL-33 in development of
therapeutics for COPD.

AUTHORS

Heather E. Raphael, Ghandi F. Hassan, Omar A. Osorio, Lucy S. Cohen, Morgan D.
Payne, Ella Katz-Kiriakos, Ishana Tata, Jamie Hicks, Derek E. Byers, Bo Zhang,
Jen Alexander-Brett

×

--------------------------------------------------------------------------------

IRON REGULATORY PROTEINS 1 AND 2 HAVE OPPOSING ROLES IN REGULATING INFLAMMATION
IN BACTERIAL ORCHITIS

Niraj Ghatpande, … , Andreas Meinhardt, Esther G. Meyron-Holtz
Niraj Ghatpande, … , Andreas Meinhardt, Esther G. Meyron-Holtz
Published February 1, 2024
Citation Information: JCI Insight. 2024;9(5):e175845.
https://doi.org/10.1172/jci.insight.175845.
View: Text | PDF



IRON REGULATORY PROTEINS 1 AND 2 HAVE OPPOSING ROLES IN REGULATING INFLAMMATION
IN BACTERIAL ORCHITIS

 * Text
 * PDF

ABSTRACT

Acute bacterial orchitis (AO) is a prevalent cause of intrascrotal inflammation,
often resulting in sub- or infertility. A frequent cause eliciting AO is
uropathogenic Escherichia coli (UPEC), a gram negative pathovar, characterized
by the expression of various iron acquisition systems to survive in a low-iron
environment. On the host side, iron is tightly regulated by iron regulatory
proteins 1 and 2 (IRP1 and -2) and these factors are reported to play a role in
testicular and immune cell function; however, their precise role remains
unclear. Here, we showed in a mouse model of UPEC-induced orchitis that the
absence of IRP1 results in less testicular damage and a reduced immune response.
Compared with infected wild-type (WT) mice, testes of UPEC-infected Irp1–/– mice
showed impaired ERK signaling. Conversely, IRP2 deletion led to a stronger
inflammatory response. Notably, differences in immune cell infiltrations were
observed among the different genotypes. In contrast with WT and Irp2–/– mice, no
increase in monocytes and neutrophils was detected in testes of Irp1–/– mice
upon UPEC infection. Interestingly, in Irp1–/– UPEC-infected testes, we observed
an increase in a subpopulation of macrophages (F4/80+CD206+) associated with
antiinflammatory and wound-healing activities compared with WT. These findings
suggest that IRP1 deletion may protect against UPEC-induced inflammation by
modulating ERK signaling and dampening the immune response.

AUTHORS

Niraj Ghatpande, Aileen Harrer, Bar Azoulay-Botzer, Noga Guttmann-Raviv,
Sudhanshu Bhushan, Andreas Meinhardt, Esther G. Meyron-Holtz

×

--------------------------------------------------------------------------------

CDK1 INHIBITION REDUCES OSTEOGENESIS IN ENDOTHELIAL CELLS IN VASCULAR
CALCIFICATION

Yan Zhao, … , Kristina I. Boström, Yucheng Yao
Yan Zhao, … , Kristina I. Boström, Yucheng Yao
Published January 23, 2024
Citation Information: JCI Insight. 2024;9(5):e176065.
https://doi.org/10.1172/jci.insight.176065.
View: Text | PDF



CDK1 INHIBITION REDUCES OSTEOGENESIS IN ENDOTHELIAL CELLS IN VASCULAR
CALCIFICATION

 * Text
 * PDF

ABSTRACT

Vascular calcification is a severe complication of cardiovascular diseases.
Previous studies demonstrated that endothelial lineage cells transitioned into
osteoblast-like cells and contributed to vascular calcification. Here, we found
that inhibition of cyclin-dependent kinase (CDK) prevented endothelial lineage
cells from transitioning to osteoblast-like cells and reduced vascular
calcification. We identified a robust induction of CDK1 in endothelial cells
(ECs) in calcified arteries and showed that EC–specific gene deletion of CDK1
decreased the calcification. We found that limiting CDK1 induced E-twenty-six
specific sequence variant 2 (ETV2), which was responsible for blocking
endothelial lineage cells from undergoing osteoblast differentiation. We also
found that inhibition of CDK1 reduced vascular calcification in a diabetic mouse
model. Together, the results highlight the importance of CDK1 suppression and
suggest CDK1 inhibition as a potential option for treating vascular
calcification.

AUTHORS

Yan Zhao, Yang Yang, Xiuju Wu, Li Zhang, Xinjiang Cai, Jaden Ji, Sydney Chen,
Abigail Vera, Kristina I. Boström, Yucheng Yao

×

--------------------------------------------------------------------------------

PURINE NUCLEOSIDE PHOSPHORYLASE INHIBITION IS AN EFFECTIVE APPROACH FOR THE
TREATMENT OF CHEMICAL HEMORRHAGIC CYSTITIS

Amanda Wolf-Johnston, … , Edwin K. Jackson, Lori A. Birder
Amanda Wolf-Johnston, … , Edwin K. Jackson, Lori A. Birder
Published January 25, 2024
Citation Information: JCI Insight. 2024;9(5):e176103.
https://doi.org/10.1172/jci.insight.176103.
View: Text | PDF



PURINE NUCLEOSIDE PHOSPHORYLASE INHIBITION IS AN EFFECTIVE APPROACH FOR THE
TREATMENT OF CHEMICAL HEMORRHAGIC CYSTITIS

 * Text
 * PDF

ABSTRACT

Hemorrhagic cystitis may be induced by infection, radiation therapy, or
medications or may be idiopathic. Along with hemorrhagic features, symptoms
include urinary urgency and frequency, dysuria (painful urination), and visceral
pain. Cystitis-induced visceral pain is one of the most challenging types of
pain to treat, and an effective treatment would address a major unmet medical
need. We assessed the efficacy of a purine nucleoside phosphorylase inhibitor,
8-aminoguanine (8-AG), for the treatment of hemorrhagic/ulcerative cystitis.
Lower urinary tract (LUT) function and structure were assessed in adult
Sprague-Dawley rats, treated chronically with cyclophosphamide (CYP; sacrificed
day 8) and randomized to daily oral treatment with 8-AG (begun 14 days prior to
CYP induction) or its vehicle. CYP-treated rats exhibited multiple
abnormalities, including increased urinary frequency and neural
mechanosensitivity, reduced bladder levels of inosine, urothelial
inflammation/damage, and activation of spinal cord microglia, which is
associated with pain hypersensitivity. 8-AG treatment of CYP-treated rats
normalized all observed histological, structural, biochemical, and physiological
abnormalities. In cystitis 8-AG improved function and reduced both pain and
inflammation likely by increasing inosine, a tissue-protective purine
metabolite. These findings demonstrate that 8-AG has translational potential for
reducing pain and preventing bladder damage in cystitis-associated LUT
dysfunctions.

AUTHORS

Amanda Wolf-Johnston, Youko Ikeda, Irina Zabbarova, Anthony J. Kanai, Sheldon
Bastacky, Robert Moldwin, Joel N.H. Stern, Edwin K. Jackson, Lori A. Birder

×

--------------------------------------------------------------------------------

RIGHT VENTRICULAR CARDIOMYOCYTE EXPANSION ACCOMPANIES CARDIAC REGENERATION IN
NEWBORN MICE AFTER LARGE LEFT VENTRICULAR INFARCTS

Tianyuan Hu, … , Michael I. Kotlikoff, Bernd K. Fleischmann
Tianyuan Hu, … , Michael I. Kotlikoff, Bernd K. Fleischmann
Published February 6, 2024
Citation Information: JCI Insight. 2024;9(5):e176281.
https://doi.org/10.1172/jci.insight.176281.
View: Text | PDF



RIGHT VENTRICULAR CARDIOMYOCYTE EXPANSION ACCOMPANIES CARDIAC REGENERATION IN
NEWBORN MICE AFTER LARGE LEFT VENTRICULAR INFARCTS

 * Text
 * PDF

ABSTRACT

Cauterization of the root of the left coronary artery (LCA) in the neonatal
heart on postnatal day 1 (P1) resulted in large, reproducible lesions of the
left ventricle (LV), and an attendant marked adaptive response in the right
ventricle (RV). The response of both chambers to LV myocardial infarction
involved enhanced cardiomyocyte (CM) division and binucleation, as well as LV
revascularization, leading to restored heart function within 7 days post surgery
(7 dps). By contrast, infarction of P3 mice resulted in cardiac scarring without
a significant regenerative and adaptive response of the LV and the RV, leading
to subsequent heart failure and death within 7 dps. The prominent RV myocyte
expansion in P1 mice involved an acute increase in pulmonary arterial pressure
and a unique gene regulatory response, leading to an increase in RV mass and
preserved heart function. Thus, distinct adaptive mechanisms in the RV, such as
CM proliferation and RV expansion, enable marked cardiac regeneration of the
infarcted LV at P1 and full functional recovery.

AUTHORS

Tianyuan Hu, Mona Malek Mohammadi, Fabian Ebach, Michael Hesse, Michael I.
Kotlikoff, Bernd K. Fleischmann

×

--------------------------------------------------------------------------------

IMPAIRED INNATE AND ADAPTIVE IMMUNE RESPONSES TO BNT162B2 SARS-COV-2 VACCINATION
IN SYSTEMIC LUPUS ERYTHEMATOSUS

Kavita Y. Sarin, … , Paul J. Utz, Lisa C. Zaba
Kavita Y. Sarin, … , Paul J. Utz, Lisa C. Zaba
Published March 8, 2024
Citation Information: JCI Insight. 2024;9(5):e176556.
https://doi.org/10.1172/jci.insight.176556.
View: Text | PDF



IMPAIRED INNATE AND ADAPTIVE IMMUNE RESPONSES TO BNT162B2 SARS-COV-2 VACCINATION
IN SYSTEMIC LUPUS ERYTHEMATOSUS

 * Text
 * PDF

ABSTRACT

Understanding the immune responses to SARS-CoV-2 vaccination is critical to
optimizing vaccination strategies for individuals with autoimmune diseases, such
as systemic lupus erythematosus (SLE). Here, we comprehensively analyzed innate
and adaptive immune responses in 19 patients with SLE receiving a complete
2-dose Pfizer-BioNTech mRNA vaccine (BNT162b2) regimen compared with a control
cohort of 56 healthy control (HC) volunteers. Patients with SLE exhibited
impaired neutralizing antibody production and antigen-specific CD4+ and CD8+ T
cell responses relative to HC. Interestingly, antibody responses were only
altered in patients with SLE treated with immunosuppressive therapies, whereas
impairment of antigen-specific CD4+ and CD8+ T cell numbers was independent of
medication. Patients with SLE also displayed reduced levels of circulating CXC
motif chemokine ligands, CXCL9, CXCL10, CXCL11, and IFN-γ after secondary
vaccination as well as downregulation of gene expression pathways indicative of
compromised innate immune responses. Single-cell RNA-Seq analysis reveals that
patients with SLE showed reduced levels of a vaccine-inducible monocyte
population characterized by overexpression of IFN-response transcription
factors. Thus, although 2 doses of BNT162b2 induced relatively robust immune
responses in patients with SLE, our data demonstrate impairment of both innate
and adaptive immune responses relative to HC, highlighting a need for
population-specific vaccination studies.

AUTHORS

Kavita Y. Sarin, Hong Zheng, Yashaar Chaichian, Prabhu S. Arunachalam, Gayathri
Swaminathan, Alec Eschholz, Fei Gao, Oliver F. Wirz, Brandon Lam, Emily Yang,
Lori W. Lee, Allan Feng, Matthew A. Lewis, Janice Lin, Holden T. Maecker, Scott
D. Boyd, Mark M. Davis, Kari C. Nadeau, Bali Pulendran, Purvesh Khatri, Paul J.
Utz, Lisa C. Zaba

×

--------------------------------------------------------------------------------

GNAS AS2 METHYLATION STATUS ENABLES MECHANISM-BASED CATEGORIZATION OF
PSEUDOHYPOPARATHYROIDISM TYPE 1B

Yorihiro Iwasaki, … , Harald Jüppner, Murat Bastepe
Yorihiro Iwasaki, … , Harald Jüppner, Murat Bastepe
Published January 30, 2024
Citation Information: JCI Insight. 2024;9(5):e177190.
https://doi.org/10.1172/jci.insight.177190.
View: Text | PDF



GNAS AS2 METHYLATION STATUS ENABLES MECHANISM-BASED CATEGORIZATION OF
PSEUDOHYPOPARATHYROIDISM TYPE 1B

 * Text
 * PDF

ABSTRACT

Pseudohypoparathyroidism type 1B (PHP1B) results from aberrant genomic
imprinting at the GNAS gene. Defining the underlying genetic cause in new
patients is challenging because various genetic alterations (e.g., deletions,
insertions) within the GNAS genomic region, including the neighboring STX16
gene, can cause PHP1B, and the genotype-epigenotype correlation has not been
clearly established. Here, by analyzing patients with PHP1B with a wide variety
of genotypes and epigenotypes, we identified a GNAS differentially methylated
region (DMR) of distinct diagnostic value. This region, GNAS AS2, was
hypomethylated in patients with genetic alterations located centromeric but not
telomeric of this DMR. The AS2 methylation status was captured by a single probe
of the methylation-sensitive multiplex ligation–dependent probe amplification
(MS-MLPA) assay utilized to diagnose PHP1B. In human embryonic stem cells, where
NESP55 transcription regulates GNAS methylation status on the maternal allele,
AS2 methylation depended on 2 imprinting control regions (STX16-ICR and
NESP-ICR) essential for NESP55 transcription. These results suggest that the AS2
methylation status in patients with PHP1B reflects the position at which the
genetic alteration affects NESP55 transcription during an early embryonic
period. Therefore, AS2 methylation levels can enable mechanistic PHP1B
categorization based on genotype-epigenotype correlation and, thus, help
identify the underlying molecular defect in patients.

AUTHORS

Yorihiro Iwasaki, Monica Reyes, Harald Jüppner, Murat Bastepe

×

--------------------------------------------------------------------------------

ENDOTHELIAL TDP-43 CONTROLS SPROUTING ANGIOGENESIS AND VASCULAR BARRIER
INTEGRITY, AND ITS DELETION TRIGGERS NEUROINFLAMMATION

Víctor Arribas, … , Bettina Schmid, Eloi Montanez
Víctor Arribas, … , Bettina Schmid, Eloi Montanez
Published February 1, 2024
Citation Information: JCI Insight. 2024;9(5):e177819.
https://doi.org/10.1172/jci.insight.177819.
View: Text | PDF



ENDOTHELIAL TDP-43 CONTROLS SPROUTING ANGIOGENESIS AND VASCULAR BARRIER
INTEGRITY, AND ITS DELETION TRIGGERS NEUROINFLAMMATION

 * Text
 * PDF

ABSTRACT

TAR DNA-binding protein 43 (TDP-43) is a DNA/RNA-binding protein that regulates
gene expression, and its malfunction in neurons has been causally associated
with multiple neurodegenerative disorders. Although progress has been made in
understanding the functions of TDP-43 in neurons, little is known about its
roles in endothelial cells (ECs), angiogenesis, and vascular function. Using
inducible EC-specific TDP-43–KO mice, we showed that TDP-43 is required for
sprouting angiogenesis, vascular barrier integrity, and blood vessel stability.
Postnatal EC-specific deletion of TDP-43 led to retinal hypovascularization due
to defects in vessel sprouting associated with reduced EC proliferation and
migration. In mature blood vessels, loss of TDP-43 disrupted the blood-brain
barrier and triggered vascular degeneration. These vascular defects were
associated with an inflammatory response in the CNS with activation of microglia
and astrocytes. Mechanistically, deletion of TDP-43 disrupted the fibronectin
matrix around sprouting vessels and reduced β-catenin signaling in ECs.
Together, our results indicate that TDP-43 is essential for the formation of a
stable and mature vasculature.

AUTHORS

Víctor Arribas, Yara Onetti, Marina Ramiro-Pareta, Pilar Villacampa, Heike Beck,
Mariona Alberola, Anna Esteve-Codina, Angelika Merkel, Markus Sperandio, Ofelia
M. Martínez-Estrada, Bettina Schmid, Eloi Montanez

×

--------------------------------------------------------------------------------

AXL INHIBITION SUPPRESSES EARLY ALLOGRAFT MONOCYTE-TO-MACROPHAGE DIFFERENTIATION
AND PROLONGS ALLOGRAFT SURVIVAL

Collin Z. Jordan, … , Edward B. Thorp, Xunrong Luo
Collin Z. Jordan, … , Edward B. Thorp, Xunrong Luo
Published January 23, 2024
Citation Information: JCI Insight. 2024;9(5):e178502.
https://doi.org/10.1172/jci.insight.178502.
View: Text | PDF



AXL INHIBITION SUPPRESSES EARLY ALLOGRAFT MONOCYTE-TO-MACROPHAGE DIFFERENTIATION
AND PROLONGS ALLOGRAFT SURVIVAL

 * Text
 * PDF

ABSTRACT

Innate immune cells are important in the initiation and potentiation of
alloimmunity in transplantation. Immediately upon organ anastomosis and
reperfusion, recipient monocytes enter the graft from circulation and
differentiate to inflammatory macrophages to promote allograft inflammation.
However, factors that drive their differentiation to inflammatory macrophages
are not understood. Here, we show that the receptor tyrosine kinase AXL was a
key driver of early intragraft differentiation of recipient infiltrating
monocytes to inflammatory macrophages in the presence of allogeneic stimulation
and cell-to-cell contact. In this context, the differentiated inflammatory
macrophages were capable of efficient alloantigen presentation and
allostimulation of T cells of the indirect pathway. Consequently, early and
transient AXL inhibition with the pharmacological inhibitor bemcentinib resulted
in a profound reduction of initial allograft inflammation and a significant
prolongation of allograft survival in a murine heart transplant model. Our
results support further investigation of AXL inhibition as part of an induction
regimen for transplantation.

AUTHORS

Collin Z. Jordan, Matthew Tunbridge, Irma Husain, Hiroki Kitai, Miriam E. Dilts,
Olivia K. Fay, Koki Abe, Catherine Xiang, Jean Kwun, Tomokazu Souma, Edward B.
Thorp, Xunrong Luo

×
Corrigendum

CIRCADIAN REGULATION OF LUNG REPAIR AND REGENERATION

Amruta Naik, … , Edward E. Morrisey, Shaon Sengupta
Amruta Naik, … , Edward E. Morrisey, Shaon Sengupta
Published March 8, 2024
Citation Information: JCI Insight. 2024;9(5):e179745.
https://doi.org/10.1172/jci.insight.179745.
View: Text | PDF | Amended Article


CIRCADIAN REGULATION OF LUNG REPAIR AND REGENERATION

 * Text
 * PDF

ABSTRACT



AUTHORS

Amruta Naik, Kaitlyn M. Forrest, Oindrila Paul, Yasmine Issah, Utham K.
Valekunja, Soon Y. Tang, Akhilesh B. Reddy, Elizabeth J. Hennessy, Thomas G.
Brooks, Fatima Chaudhry, Apoorva Babu, Michael Morley, Jarod A. Zepp, Gregory R.
Grant, Garret A. FitzGerald, Amita Sehgal, G. Scott Worthen, David B. Frank,
Edward E. Morrisey, Shaon Sengupta

×

--------------------------------------------------------------------------------

IN-PRESS PREVIEW - MORE

INTEGRATED PLASMA PROTEOMICS IDENTIFIES TUBERCULOSIS-SPECIFIC DIAGNOSTIC
BIOMARKERS

Novel biomarkers to identify infectious patients transmitting Mycobacterium
tuberculosis are urgently needed to control the global tuberculosis (TB)
pandemic. We hypothesized that proteins released...
Published March 21, 2024

Clinical Medicine In-Press Preview Infectious disease Pulmonology

INTEGRATED PLASMA PROTEOMICS IDENTIFIES TUBERCULOSIS-SPECIFIC DIAGNOSTIC
BIOMARKERS

 * Text
 * PDF

ABSTRACT

Novel biomarkers to identify infectious patients transmitting Mycobacterium
tuberculosis are urgently needed to control the global tuberculosis (TB)
pandemic. We hypothesized that proteins released into the plasma in active
pulmonary TB are clinically useful biomarkers to distinguish TB cases from
healthy individuals and patients with other respiratory infections. We applied a
highly sensitive non-depletion tandem mass spectrometry discovery approach to
investigate plasma protein expression in pulmonary TB cases compared to healthy
controls in South African and Peruvian cohorts. Bioinformatic analysis using
linear modelling and network correlation analyses identified 118 differentially
expressed proteins, significant through three complementary analytical
pipelines. Candidate biomarkers were subsequently analysed in two validation
cohorts of differing ethnicity using antibody-based proximity extension assays.
TB-specific host biomarkers were confirmed. A six-protein diagnostic panel,
comprising FETUB, FCGR3B, LRG1, SELL, CD14 and ADA2, differentiated patients
with pulmonary TB from healthy controls and patients with other respiratory
infections with high sensitivity and specificity in both cohorts. This biomarker
panel exceeds the World Health Organisation Target Product Profile specificity
criteria for a triage test for TB. The new biomarkers have potential for further
development as near-patient TB screening assays, thereby helping to close the
case-detection gap that fuels the global pandemic.

AUTHORS

Hannah F. Schiff, Naomi F. Walker, Cesar Ugarte-Gil, Marc Tebruegge, Antigoni
Manousopoulou, Spiros D. Garbis, Salah Mansour, Pak Ho Wong, Gabrielle Rockett,
Paolo Piazza, Mahesan Niranjan, Andres F. Vallejo, Christopher H. Woelk, Robert
J. Wilkinson, Liku B. Tezera, Diana Garay-Baquero, Paul Elkington

×

--------------------------------------------------------------------------------

TUMOR TREATING FIELDS SUPPRESS TUMOR CELL GROWTH AND NEUROLOGIC DECLINE IN
MODELS OF SPINE METASTASES

Spine metastases can result in severe neurologic compromise and decreased
overall survival. Despite treatment advances, local disease progression is
frequent, highlighting the need for novel...
Published March 21, 2024

Resource and Technical Advance In-Press Preview Bone biology Neuroscience

TUMOR TREATING FIELDS SUPPRESS TUMOR CELL GROWTH AND NEUROLOGIC DECLINE IN
MODELS OF SPINE METASTASES

 * Text
 * PDF

ABSTRACT

Spine metastases can result in severe neurologic compromise and decreased
overall survival. Despite treatment advances, local disease progression is
frequent, highlighting the need for novel therapies. Tumor treating fields
(TTFields) impair tumor cell replication and are influenced by properties of
surrounding tissue. We hypothesize bone’s dielectric properties will enhance
TTFields mediated suppression of tumor growth in spine metastasis models.
Computational modeling of TTFields intensity was performed following surgical
resection of a spinal metastasis and demonstrated enhanced TTFields intensity
within the resected vertebral body. Additionally, luciferase-tagged human KRIB
osteosarcoma and A549 lung adenocarcinoma cell lines were cultured in
demineralized bone grafts and exposed to TTFields. Following TTFields exposure,
BLI signal decreased 10-80% of baseline while control cultures displayed
4.48-9.36 fold increase in signal. Lastly, TTFields were applied in an
orthotopic murine model of spinal metastasis. After 21 days of treatment,
control mice demonstrated a 5-fold increase in BLI signal compared to TTFields
treated mice. TTFields similarly prevented tumor invasion into the spinal canal
and development of neurologic symptoms. Our data suggest that TTFields can be
leveraged as a local therapy within minimally-conductive bone of spine
metastases. This provides the groundwork for future studies investigating
TTFields for patients with treatment-refractory spine metastases.

AUTHORS

Daniel Ledbetter, Romulo de Almeida, Xizi Wu, Ariel Naveh, Chirag B. Patel,
Queena Gonzalez, Thomas H. Beckham, Robert North, Laurence Rhines, Jing Li, Amol
Ghia, David Aten, Claudio Tatsui, Christopher Alvarez-Breckenridge

×

--------------------------------------------------------------------------------

HIPK2 C-TERMINAL DOMAIN INHIBITS NF-ΚB SIGNALING AND RENAL INFLAMMATION IN
KIDNEY INJURY

HIPK2 is a multifunctional kinase that acts as a key pathogenic mediator of
chronic kidney disease and fibrosis. It acts as a central effector of multiple
signaling pathways implicated in kidney...
Published March 21, 2024

Research In-Press Preview Nephrology

HIPK2 C-TERMINAL DOMAIN INHIBITS NF-ΚB SIGNALING AND RENAL INFLAMMATION IN
KIDNEY INJURY

 * Text
 * PDF

ABSTRACT

HIPK2 is a multifunctional kinase that acts as a key pathogenic mediator of
chronic kidney disease and fibrosis. It acts as a central effector of multiple
signaling pathways implicated in kidney injury, such as TGF-β/Smad3-mediated
extracellular matrix accumulation, NF-κB-mediated inflammation, and p53-mediated
apoptosis. Thus, a better understanding of the specific HIPK2 regions necessary
for distinct downstream pathway activation is critical for optimal drug
development for CKD. Our study now shows that Caspase 6-mediated removal of the
C-terminal region of HIPK2 (HIPK2-CT) leads to hyperactive p65 NF-κB
transcriptional response in kidney cells. In contrast, the expression of cleaved
HIPK2-CT fragment can restrain p65 NF-κB transcriptional activity by cytoplasmic
sequestration NF-κB signaling component, p65 NF-κB, and attenuation of IκBα
degradation. Therefore, we examined whether HIPK2-CT expression can be exploited
to restrain renal inflammation in vivo. The induction of HIPK2-CT overexpression
in kidney tubular cells attenuated p65 nuclear translocation, expression of
inflammatory cytokines, and macrophage infiltration in the kidney of mice with
unilateral ureteral obstruction and lipopolysaccharide-induced acute kidney
injury. Collectively, our findings indicate that the C-terminal region of HIPK2
is involved in the regulation of nuclear NF-κB transcriptional activity and that
HIPK2-CT or its analogs could be further exploited as potential
anti-inflammatory agents to treat kidney disease.

AUTHORS

Ye Feng, Zhengzhe Li, Heather Wang, Bi-Cheng Liu, Kyung Lee, John Cijiang He

×

--------------------------------------------------------------------------------

MAPPING SCA1 REGIONAL VULNERABILITIES REVEALS NEURAL AND SKELETAL MUSCLE
CONTRIBUTIONS TO DISEASE

Spinocerebellar ataxia type 1 (SCA1) is a fatal neurodegenerative disease caused
by an expanded polyglutamine tract in the widely expressed ataxin-1 (ATXN1)
protein. To elucidate anatomical regions...
Published March 21, 2024

Research In-Press Preview Neuroscience

MAPPING SCA1 REGIONAL VULNERABILITIES REVEALS NEURAL AND SKELETAL MUSCLE
CONTRIBUTIONS TO DISEASE

 * Text
 * PDF

ABSTRACT

Spinocerebellar ataxia type 1 (SCA1) is a fatal neurodegenerative disease caused
by an expanded polyglutamine tract in the widely expressed ataxin-1 (ATXN1)
protein. To elucidate anatomical regions and cell types that underlie mutant
ATXN1-induced disease phenotypes, we developed a floxed conditional knockin
mouse (f-ATXN1146Q/2Q) with mouse Atxn1 coding exons replaced by human ATXN1
exons encoding 146 glutamines. f-ATXN1146Q/2Q mice manifested SCA1-like
phenotypes including motor and cognitive deficits, wasting, and decreased
survival. Central nervous system (CNS) contributions to disease were revealed
using f-ATXN1146Q/2Q;Nestin-Cre mice, that showed improved rotarod, open field,
and Barnes maze performance by 6-12 weeks-of-age. In contrast, striatal
contributions to motor deficits using f-ATXN1146Q/2Q;Rgs9-Cre mice revealed that
mice lacking ATXN1146Q/2Q in striatal medium-spiny neurons showed a trending
improvement in rotarod performance at 30 weeks-of-age. Surprisingly, a prominent
role for muscle contributions to disease was revealed in
f-ATXN1146Q/2Q;ACTA1-Cre mice based on their recovery from kyphosis and absence
of muscle pathology. Collectively, data from the targeted conditional deletion
of the expanded allele demonstrated CNS and peripheral contributions to disease
and highlighted the need to consider muscle in addition to the brain for optimal
SCA1 therapeutics.

AUTHORS

Lisa Duvick, W. Michael Southern, Kellie A. Benzow, Zoe N. Burch, Hillary P.
Handler, Jason S. Mitchell, Hannah Kuivinen, Udaya Gadiparthi, Praseuth Yang,
Alyssa Soles, Carrie A. Sheeler, Orion Rainwater, Shannah Serres, Erin B. Lind,
Tessa Nichols-Meade, Brennon O'Callaghan, Huda Y. Zoghbi, Marija Cvetanovic,
Vanessa C. Wheeler, James M. Ervasti, Michael D. Koob, Harry T. Orr

×

--------------------------------------------------------------------------------

EVALUATING IMMUNOTHERAPEUTIC OUTCOMES IN TRIPLE NEGATIVE BREAST CANCER WITH A
CHOLESTEROL RADIOTRACER IN MICE

Evaluating the response to immune checkpoint inhibitors (ICIs) remains an unmet
challenge in triple-negative breast cancer (TNBC). The requirement of
cholesterol for activation and function of T...
Published March 19, 2024

Resource and Technical Advance In-Press Preview Immunology Oncology

EVALUATING IMMUNOTHERAPEUTIC OUTCOMES IN TRIPLE NEGATIVE BREAST CANCER WITH A
CHOLESTEROL RADIOTRACER IN MICE

 * Text
 * PDF

ABSTRACT

Evaluating the response to immune checkpoint inhibitors (ICIs) remains an unmet
challenge in triple-negative breast cancer (TNBC). The requirement of
cholesterol for activation and function of T cells led us to hypothesize that
quantifying cellular accumulation of this molecule could distinguish successful
from ineffective checkpoint immunotherapy. To analyze accumulation of
cholesterol by T cells in the immune microenvironment of breast cancer, we
leveraged the PET radiotracer, eFNP-59. eFNP-59 is an analog of cholesterol that
our group validated as an imaging biomarker for cholesterol uptake in
pre-clinical models and initial human studies. In immunocompetent mouse models
of TNBC, we found that elevated uptake of exogenous labeled cholesterol analogs
functions as a marker for T cell activation. When comparing ICI-responsive and
non-responsive tumors directly, uptake of fluorescent cholesterol and eFNP-59
increased in T cells from ICI-responsive tumors. We discovered that accumulation
of cholesterol by T cells increased in ICI-responding tumors that received
anti-PD-1 checkpoint immunotherapy. In patients with TNBC, tumors containing
cycling T cells had features of cholesterol uptake and trafficking within those
populations. These results suggest that uptake of exogenous cholesterol analogs
by tumor-infiltrating T cells detects T cell activation and has potential to
assess the success of ICI therapy.

AUTHORS

Nicholas G. Ciavattone, Jenny Nan Guan, Alex Farfel, Jenelle Stauff, Timothy J.
Desmond, Benjamin L. Viglianti, Peter J.H. Scott, Allen F. Brooks, Gary D. Luker

×

--------------------------------------------------------------------------------

VIEW MORE ARTICLES BY TOPIC:

Aging

AIDS/HIV

Angiogenesis

Autoimmunity

Bone biology

Cardiology

Cell biology

Clinical trials

COVID-19

Dermatology

Development

Endocrinology

Gastroenterology

Genetics

Hematology

Hepatology

Immunology

Infectious disease

Inflammation

Metabolism

Microbiology

Muscle biology

Nephrology

Neuroscience

Oncology

Ophthalmology

Otology

Pulmonology

Reproductive biology

Stem cells

Therapeutics

Transplantation

Vaccines

Vascular biology

Virology

Advertisement


SIGN UP FOR EMAIL ALERTS



LATEST REVIEWS - MORE

THE IMMUNE SYSTEM AND METABOLIC PRODUCTS IN EPILEPSY AND GLIOMA-ASSOCIATED
EPILEPSY: EMERGING THERAPEUTIC DIRECTIONS

Epilepsy has a profound impact on quality of life. Despite the development of
new antiseizure medications (ASMs), approximately one-third of affected patients
have drug-refractory epilepsy and are...

MUSCLE: AN INDEPENDENT CONTRIBUTOR TO THE NEUROMUSCULAR SPINAL MUSCULAR ATROPHY
DISEASE PHENOTYPE

Spinal muscular atrophy (SMA) is a pediatric-onset neuromuscular disorder caused
by insufficient survival motor neuron (SMN) protein. SMN restorative therapies
are now approved for the treatment of...

AUTHOR'S TAKE - MORE

QUISINOSTAT AS A RADIOSENSITIZER FOR THE TREATMENT OF GLIOBLASTOMA

In this episode, Costanza Lo Cascio, Artak Tovmasyan, and Shwetal Mehta discuss
the identification of a brain-penetrant HDAC inhibitor with radiosensitizing
properties for the treatment of glioblastoma...




Copyright © 2024 American Society for Clinical Investigation
ISSN 2379-3708

SIGN UP FOR EMAIL ALERTS


Posted by 6 X users
3 readers on Mendeley
See more details
Posted by 1 X users
See more details
Posted by 3 X users
See more details
Posted by 25 X users
See more details
Picked up by 8 news outlets
Blogged by 1
Posted by 14 X users
7 readers on Mendeley
See more details
Posted by 5 X users
See more details
Posted by 13 X users
2 readers on Mendeley
See more details
Posted by 5 X users
See more details
Posted by 1 X users
2 readers on Mendeley
See more details
Posted by 1 X users
See more details
Posted by 10 X users
See more details
Posted by 10 X users
See more details
Posted by 3 X users
1 readers on Mendeley
See more details
Posted by 1 X users
See more details
Posted by 2 X users
See more details
Posted by 3 X users
See more details
Picked up by 4 news outlets
Blogged by 1
Posted by 2 X users
See more details
Posted by 3 X users
1 readers on Mendeley
See more details
Posted by 1 X users
See more details
Picked up by 1 news outlets
Posted by 13 X users
2 readers on Mendeley
See more details
Posted by 15 X users
1 readers on Mendeley
See more details
Posted by 14 X users
1 readers on Mendeley
See more details
Posted by 1 X users
See more details