Go to The Journal of Clinical Investigation
  • About
  • Editors
  • Consulting Editors
  • For authors
  • Publication ethics
  • Transfers
  • Advertising
  • Job board
  • Contact
  • Current issue
  • Past issues
  • By specialty
    • COVID-19
    • Cardiology
    • Immunology
    • Metabolism
    • Nephrology
    • Oncology
    • Pulmonology
    • All ...
  • Videos
  • Collections
    • Resource and Technical Advances
    • Clinical Medicine
    • Reviews
    • Editorials
    • Perspectives
    • Top read articles
  • JCI This Month
    • Current issue
    • Past issues

  • Current issue
  • Past issues
  • Specialties
  • In-Press Preview
  • Editorials
  • Viewpoint
  • Top read articles
  • About
  • Editors
  • Consulting Editors
  • For authors
  • Publication ethics
  • Transfers
  • Advertising
  • Job board
  • Contact

  • 2,893 Articles
  • 0 Posts
  • ← Previous
  • 1
  • 2
  • …
  • 281
  • 282
  • 283
  • …
  • 289
  • 290
  • Next →
The MEK inhibitor trametinib separates murine graft-versus-host disease from graft-versus-tumor effects
Hidekazu Itamura, … , Krishna V. Komanduri, Shinya Kimura
Hidekazu Itamura, … , Krishna V. Komanduri, Shinya Kimura
Published July 7, 2016
Citation Information: JCI Insight. 2016;1(10):e86331. https://doi.org/10.1172/jci.insight.86331.
View: Text | PDF

The MEK inhibitor trametinib separates murine graft-versus-host disease from graft-versus-tumor effects

  • Text
  • PDF
Abstract

The efficacy of allogeneic hematopoietic stem cell transplantation for hematologic malignancies is limited by the difficulty in suppressing graft-versus-host disease (GVHD) without compromising graft-versus-tumor (GVT) effects. We previously showed that RAS/MEK/ERK signaling depends on memory differentiation in human T cells, which confers susceptibility to selective inhibition of naive T cells. Actually, antineoplastic MEK inhibitors selectively suppress alloreactive T cells, sparing virus-specific T cells in vitro. Here, we show that trametinib, a MEK inhibitor clinically approved for melanoma, suppresses GVHD safely without affecting GVT effects in vivo. Trametinib prolonged survival of GVHD mice and attenuated GVHD symptoms and pathology in the gut and skin. It inhibited ERK1/2 phosphorylation and expansion of donor T cells, sparing Tregs and B cells. Although high-dose trametinib inhibited myeloid cell engraftment, low-dose trametinib suppressed GVHD without severe adverse events. Notably, trametinib facilitated the survival of mice transplanted with allogeneic T cells and P815 tumor cells with no residual P815 cells observed in the livers and spleens, whereas tacrolimus resulted in P815 expansion. These results confirm that trametinib selectively suppresses GVHD-inducing T cells while sparing antitumor T cells in vivo, which makes it a promising candidate for translational studies aimed at preventing or treating GVHD.

Authors

Hidekazu Itamura, Takero Shindo, Isao Tawara, Yasushi Kubota, Ryusho Kariya, Seiji Okada, Krishna V. Komanduri, Shinya Kimura

×

Deep sequencing reveals microRNAs predictive of antiangiogenic drug response
Jesús García-Donas, … , Mercedes Robledo, Cristina Rodriguez-Antona
Jesús García-Donas, … , Mercedes Robledo, Cristina Rodriguez-Antona
Published July 7, 2016
Citation Information: JCI Insight. 2016;1(10):e86051. https://doi.org/10.1172/jci.insight.86051.
View: Text | PDF

Deep sequencing reveals microRNAs predictive of antiangiogenic drug response

  • Text
  • PDF
Abstract

The majority of metastatic renal cell carcinoma (RCC) patients are treated with tyrosine kinase inhibitors (TKI) in first-line treatment; however, a fraction are refractory to these antiangiogenic drugs. MicroRNAs (miRNAs) are regulatory molecules proven to be accurate biomarkers in cancer. Here, we identified miRNAs predictive of progressive disease under TKI treatment through deep sequencing of 74 metastatic clear cell RCC cases uniformly treated with these drugs. Twenty-nine miRNAs were differentially expressed in the tumors of patients who progressed under TKI therapy (P values from 6 × 10–9 to 3 × 10–3). Among 6 miRNAs selected for validation in an independent series, the most relevant associations corresponded to miR–1307-3p, miR–155-5p, and miR–221-3p (P = 4.6 × 10–3, 6.5 × 10–3, and 3.4 × 10–2, respectively). Furthermore, a 2 miRNA–based classifier discriminated individuals with progressive disease upon TKI treatment (AUC = 0.75, 95% CI, 0.64–0.85; P = 1.3 × 10–4) with better predictive value than clinicopathological risk factors commonly used. We also identified miRNAs significantly associated with progression-free survival and overall survival (P = 6.8 × 10–8 and 7.8 × 10–7 for top hits, respectively), and 7 overlapped with early progressive disease. In conclusion, this is the first miRNome comprehensive study, to our knowledge, that demonstrates a predictive value of miRNAs for TKI response and provides a new set of relevant markers that can help rationalize metastatic RCC treatment.

Authors

Jesús García-Donas, Benoit Beuselinck, Lucía Inglada-Pérez, Osvaldo Graña, Patrick Schöffski, Agnieszka Wozniak, Oliver Bechter, Maria Apellániz-Ruiz, Luis Javier Leandro-García, Emilio Esteban, Daniel E. Castellano, Aranzazu González del Alba, Miguel Angel Climent, Susana Hernando, José Angel Arranz, Manuel Morente, David G. Pisano, Mercedes Robledo, Cristina Rodriguez-Antona

×

An extra copy of p53 suppresses development of spontaneous Kras-driven but not radiation-induced cancer
Everett J. Moding, … , Chang-Lung Lee, David G. Kirsch
Everett J. Moding, … , Chang-Lung Lee, David G. Kirsch
Published July 7, 2016
Citation Information: JCI Insight. 2016;1(10):e86698. https://doi.org/10.1172/jci.insight.86698.
View: Text | PDF

An extra copy of p53 suppresses development of spontaneous Kras-driven but not radiation-induced cancer

  • Text
  • PDF
Abstract

The tumor suppressor p53 blocks tumor progression in multiple tumor types. Radiation-induced cancer following exposure to radiation therapy or space travel may also be regulated by p53 because p53 has been proposed to respond to DNA damage to suppress tumorigenesis. Here, we investigate the role of p53 in lung carcinogenesis and lymphomagenesis in LA-1 KrasG12D mice with wild-type p53 or an extra copy of p53 (super p53) exposed to fractionated total body irradiation with low linear energy transfer (low-LET) X-rays or high-LET iron ions and compared tumor formation in these mice with unirradiated controls. We found that an additional copy of p53 suppressed both Kras-driven lung tumor and lymphoma development in the absence of radiation. However, an additional copy of p53 did not affect lymphoma development following low- or high-LET radiation exposure and was unable to suppress radiation-induced expansion of thymocytes with mutated Kras. Moreover, radiation exposure increased lung tumor size in super p53 but not wild-type p53 mice. These results demonstrate that although p53 suppresses the development of spontaneous tumors expressing KrasG12D, in the context of exposure to ionizing radiation, an extra copy of p53 does not protect against radiation-induced lymphoma and may promote KrasG12D mutant lung cancer.

Authors

Everett J. Moding, Hooney D. Min, Katherine D. Castle, Moiez Ali, Loretta Woodlief, Nerissa Williams, Yan Ma, Yongbaek Kim, Chang-Lung Lee, David G. Kirsch

×

Prohibitin/annexin 2 interaction regulates fatty acid transport in adipose tissue
Ahmad Salameh, … , Wadih Arap, Mikhail G. Kolonin
Ahmad Salameh, … , Wadih Arap, Mikhail G. Kolonin
Published July 7, 2016
Citation Information: JCI Insight. 2016;1(10):e86351. https://doi.org/10.1172/jci.insight.86351.
View: Text | PDF

Prohibitin/annexin 2 interaction regulates fatty acid transport in adipose tissue

  • Text
  • PDF
Abstract

We have previously identified prohibitin (PHB) and annexin A2 (ANX2) as proteins interacting on the surface of vascular endothelial cells in white adipose tissue (WAT) of humans and mice. Here, we demonstrate that ANX2 and PHB also interact in adipocytes. Mice lacking ANX2 have normal WAT vascularization, adipogenesis, and glucose metabolism but display WAT hypotrophy due to reduced fatty acid uptake by WAT endothelium and adipocytes. By using cell culture systems in which ANX2/PHB binding is disrupted either genetically or through treatment with a blocking peptide, we show that fatty acid transport efficiency relies on this protein complex. We also provide evidence that the interaction between ANX2 and PHB mediates fatty acid transport from the endothelium into adipocytes. Moreover, we demonstrate that ANX2 and PHB form a complex with the fatty acid transporter CD36. Finally, we show that the colocalization of PHB and CD36 on adipocyte surface is induced by extracellular fatty acids. Together, our results suggest that an unrecognized biochemical interaction between ANX2 and PHB regulates CD36-mediated fatty acid transport in WAT, thus revealing a new potential pathway for intervention in metabolic diseases.

Authors

Ahmad Salameh, Alexes C. Daquinag, Daniela I. Staquicini, Zhiqiang An, Katherine A. Hajjar, Renata Pasqualini, Wadih Arap, Mikhail G. Kolonin

×

Induced regulatory T cells in allograft tolerance via transient mixed chimerism
Kiyohiko Hotta, … , A. Benedict Cosimi, Tatsuo Kawai
Kiyohiko Hotta, … , A. Benedict Cosimi, Tatsuo Kawai
Published July 7, 2016
Citation Information: JCI Insight. 2016;1(10):e86419. https://doi.org/10.1172/jci.insight.86419.
View: Text | PDF

Induced regulatory T cells in allograft tolerance via transient mixed chimerism

  • Text
  • PDF
Abstract

Successful induction of allograft tolerance has been achieved in nonhuman primates (NHPs) and humans via induction of transient hematopoietic chimerism. Since allograft tolerance was achieved in these recipients without durable chimerism, peripheral mechanisms are postulated to play a major role. Here, we report our studies of T cell immunity in NHP recipients that achieved long-term tolerance versus those that rejected the allograft (AR). All kidney, heart, and lung transplant recipients underwent simultaneous or delayed donor bone marrow transplantation (DBMT) following conditioning with a nonmyeloablative regimen. After DBMT, mixed lymphocyte culture with CFSE consistently revealed donor-specific loss of CD8+ T cell responses in tolerant (TOL) recipients, while marked CD4+ T cell proliferation in response to donor antigens was found to persist. Interestingly, a significant proportion of the proliferated CD4+ cells were FOXP3+ in TOL recipients, but not in AR or naive NHPs. In TOL recipients, CD4+FOXP3+ cell proliferation against donor antigens was greater than that observed against third-party antigens. Finally, the expanded Tregs appeared to be induced Tregs (iTregs) that were converted from non-Tregs. These data provide support for the hypothesis that specific induction of iTregs by donor antigens is key to long-term allograft tolerance induced by transient mixed chimerism.

Authors

Kiyohiko Hotta, Akihiro Aoyama, Tetsu Oura, Yohei Yamada, Makoto Tonsho, Kyu Ha Huh, Kento Kawai, David Schoenfeld, James S. Allan, Joren C. Madsen, Gilles Benichou, Rex-Neal Smith, Robert B. Colvin, David H. Sachs, A. Benedict Cosimi, Tatsuo Kawai

×

PD-1 blockade enhances the vaccination-induced immune response in glioma
Joseph P. Antonios, … , Linda M. Liau, Robert M. Prins
Joseph P. Antonios, … , Linda M. Liau, Robert M. Prins
Published July 7, 2016
Citation Information: JCI Insight. 2016;1(10):e87059. https://doi.org/10.1172/jci.insight.87059.
View: Text | PDF

PD-1 blockade enhances the vaccination-induced immune response in glioma

  • Text
  • PDF
Abstract

DC vaccination with autologous tumor lysate has demonstrated promising results for the treatment of glioblastoma (GBM) in preclinical and clinical studies. While the vaccine appears capable of inducing T cell infiltration into tumors, the effectiveness of active vaccination in progressively growing tumors is less profound. In parallel, a number of studies have identified negative costimulatory pathways, such as programmed death 1/programmed death ligand 1 (PD-1/PD-L1), as relevant mediators of the intratumoral immune responses. Clinical responses to PD-1 pathway inhibition, however, have also been varied. To evaluate the relevance to established glioma, the effects of PD-1 blockade following DC vaccination were tested in intracranial (i.c.) glioma tumor–bearing mice. Treatment with both DC vaccination and PD-1 mAb blockade resulted in long-term survival, while neither agent alone induced a survival benefit in animals with larger, established tumors. This survival benefit was completely dependent on CD8+ T cells. Additionally, DC vaccine plus PD-1 mAb blockade resulted in the upregulation of integrin homing and immunologic memory markers on tumor-infiltrating lymphocytes (TILs). In clinical samples, DC vaccination in GBM patients was associated with upregulation of PD-1 expression in vivo, while ex vivo blockade of PD-1 on freshly isolated TILs dramatically enhanced autologous tumor cell cytolysis. These findings strongly suggest that the PD-1/PD-L1 pathway plays an important role in the adaptive immune resistance of established GBM in response to antitumor active vaccination and provide us with a rationale for the clinical translation of this combination therapy.

Authors

Joseph P. Antonios, Horacio Soto, Richard G. Everson, Joey Orpilla, Diana Moughon, Namjo Shin, Shaina Sedighim, William H. Yong, Gang Li, Timothy F. Cloughesy, Linda M. Liau, Robert M. Prins

×

Biofilm in group A streptococcal necrotizing soft tissue infections
Nikolai Siemens, … , Mattias Svensson, Anna Norrby-Teglund
Nikolai Siemens, … , Mattias Svensson, Anna Norrby-Teglund
Published July 7, 2016
Citation Information: JCI Insight. 2016;1(10):e87882. https://doi.org/10.1172/jci.insight.87882.
View: Text | PDF

Biofilm in group A streptococcal necrotizing soft tissue infections

  • Text
  • PDF
Abstract

Necrotizing fasciitis caused by group A streptococcus (GAS) is a life-threatening, rapidly progressing infection. At present, biofilm is not recognized as a potential problem in GAS necrotizing soft tissue infections (NSTI), as it is typically linked to chronic infections or associated with foreign devices. Here, we present a case of a previously healthy male presenting with NSTI caused by GAS. The infection persisted over 24 days, and the surgeon documented the presence of a “thick layer biofilm” in the fascia. Subsequent analysis of NSTI patient tissue biopsies prospectively included in a multicenter study revealed multiple areas of biofilm in 32% of the patients studied. Biopsies associated with biofilm formation were characterized by massive bacterial load, a pronounced inflammatory response, and clinical signs of more severe tissue involvement. In vitro infections of a human skin tissue model with GAS NSTI isolates also revealed multilayered fibrous biofilm structures, which were found to be under the control of the global Nra gene regulator. The finding of GAS biofilm formation in NSTIs emphasizes the urgent need for biofilm to be considered as a potential complicating microbiological feature of GAS NSTI and, consequently, emphasizes reconsideration of antibiotic treatment protocols.

Authors

Nikolai Siemens, Bhavya Chakrakodi, Srikanth Mairpady Shambat, Marina Morgan, Helena Bergsten, Ole Hyldegaard, Steinar Skrede, Per Arnell, Martin B. Madsen, Linda Johansson, INFECT Study Group, Julius Juarez, Lidija Bosnjak, Matthias Mörgelin, Mattias Svensson, Anna Norrby-Teglund

×

Dual epithelial and immune cell function of Dvl1 regulates gut microbiota composition and intestinal homeostasis
Haim Belinson, … , Richard M. Locksley, Ophir D. Klein
Haim Belinson, … , Richard M. Locksley, Ophir D. Klein
Published July 7, 2016
Citation Information: JCI Insight. 2016;1(10):e85395. https://doi.org/10.1172/jci.insight.85395.
View: Text | PDF

Dual epithelial and immune cell function of Dvl1 regulates gut microbiota composition and intestinal homeostasis

  • Text
  • PDF
Abstract

Homeostasis of the gastrointestinal (GI) tract is controlled by complex interactions between epithelial and immune cells and the resident microbiota. Here, we studied the role of Wnt signaling in GI homeostasis using Disheveled 1 knockout (Dvl1–/–) mice, which display an increase in whole gut transit time. This phenotype is associated with a reduction and mislocalization of Paneth cells and an increase in CD8+ T cells in the lamina propria. Bone marrow chimera experiments demonstrated that GI dysfunction requires abnormalities in both epithelial and immune cells. Dvl1–/– mice exhibit a significantly distinct GI microbiota, and manipulation of the gut microbiota in mutant mice rescued the GI transit abnormality without correcting the Paneth and CD8+ T cell abnormalities. Moreover, manipulation of the gut microbiota in wild-type mice induced a GI transit abnormality akin to that seen in Dvl1–/– mice. Together, these data indicate that microbiota manipulation can overcome host dysfunction to correct GI transit abnormalities. Our findings illustrate a mechanism by which the epithelium and immune system coregulate gut microbiota composition to promote normal GI function.

Authors

Haim Belinson, Adam K. Savage, Douglas Fadrosh, Yien-Ming Kuo, Din Lin, Ricardo Valladares, Ysbrand Nusse, Anthony Wynshaw-Boris, Susan V. Lynch, Richard M. Locksley, Ophir D. Klein

×

Mutant p53 regulates ovarian cancer transformed phenotypes through autocrine matrix deposition
Marcin P. Iwanicki, … , Ronny Drapkin, Joan S. Brugge
Marcin P. Iwanicki, … , Ronny Drapkin, Joan S. Brugge
Published July 7, 2016
Citation Information: JCI Insight. 2016;1(10):e86829. https://doi.org/10.1172/jci.insight.86829.
View: Text | PDF

Mutant p53 regulates ovarian cancer transformed phenotypes through autocrine matrix deposition

  • Text
  • PDF
Abstract

High-grade serous ovarian carcinoma (HGS-OvCa) harbors p53 mutations and can originate from the epithelial cell compartment of the fallopian tube fimbriae. From this site, neoplastic cells detach, survive in the peritoneal cavity, and form cellular clusters that intercalate into the mesothelium to form ovarian and peritoneal masses. To examine the contribution of mutant p53 to phenotypic alterations associated with HGS-OvCA, we developed live-cell microscopy assays that recapitulate these early events in cultured fallopian tube nonciliated epithelial (FNE) cells. Expression of stabilizing mutant variants of p53, but not depletion of endogenous wild-type p53, in FNE cells promoted survival and cell-cell aggregation under conditions of cell detachment, leading to the formation of cell clusters with mesothelium-intercalation capacity. Mutant p53R175H-induced phenotypes were dependent on fibronectin production, α5β1 fibronectin receptor engagement, and TWIST1 expression. These results indicate that FNE cells expressing stabilizing p53 mutants acquire anchorage independence and subsequent mesothelial intercalation capacity through a mechanism involving mesenchymal transition and matrix production. These findings provide important new insights into activities of mutant p53 in the cells of origin of HGS-OvCa.

Authors

Marcin P. Iwanicki, Hsing-Yu Chen, Claudia Iavarone, Ioannis K. Zervantonakis, Taru Muranen, Marián Novak, Tan A. Ince, Ronny Drapkin, Joan S. Brugge

×

Profiling cancer testis antigens in non–small-cell lung cancer
Dijana Djureinovic, … , Mathias Uhlén, Patrick Micke
Dijana Djureinovic, … , Mathias Uhlén, Patrick Micke
Published July 7, 2016
Citation Information: JCI Insight. 2016;1(10):e86837. https://doi.org/10.1172/jci.insight.86837.
View: Text | PDF

Profiling cancer testis antigens in non–small-cell lung cancer

  • Text
  • PDF
Abstract

Cancer testis antigens (CTAs) are of clinical interest as biomarkers and present valuable targets for immunotherapy. To comprehensively characterize the CTA landscape of non–small-cell lung cancer (NSCLC), we compared RNAseq data from 199 NSCLC tissues to the normal transcriptome of 142 samples from 32 different normal organs. Of 232 CTAs currently annotated in the Caner Testis Database (CTdatabase), 96 were confirmed in NSCLC. To obtain an unbiased CTA profile of NSCLC, we applied stringent criteria on our RNAseq data set and defined 90 genes as CTAs, of which 55 genes were not annotated in the CTdatabase, thus representing potential new CTAs. Cluster analysis revealed that CTA expression is histology dependent and concurrent expression is common. IHC confirmed tissue-specific protein expression of selected new CTAs (TKTL1, TGIF2LX, VCX, and CXORF67). Furthermore, methylation was identified as a regulatory mechanism of CTA expression based on independent data from The Cancer Genome Atlas. The proposed prognostic impact of CTAs in lung cancer was not confirmed, neither in our RNAseq cohort nor in an independent meta-analysis of 1,117 NSCLC cases. In summary, we defined a set of 90 reliable CTAs, including information on protein expression, methylation, and survival association. The detailed RNAseq catalog can guide biomarker studies and efforts to identify targets for immunotherapeutic strategies.

Authors

Dijana Djureinovic, Björn M. Hallström, Masafumi Horie, Johanna Sofia Margareta Mattsson, Linnea La Fleur, Linn Fagerberg, Hans Brunnström, Cecilia Lindskog, Katrin Madjar, Jörg Rahnenführer, Simon Ekman, Elisabeth Ståhle, Hirsh Koyi, Eva Brandén, Karolina Edlund, Jan G. Hengstler, Mats Lambe, Akira Saito, Johan Botling, Fredrik Pontén, Mathias Uhlén, Patrick Micke

×
  • ← Previous
  • 1
  • 2
  • …
  • 281
  • 282
  • 283
  • …
  • 289
  • 290
  • Next →

No posts were found with this tag.

Advertisement

Copyright © 2023 American Society for Clinical Investigation
ISSN 2379-3708

Sign up for email alerts