Go to JCI Insight
Jci spelled out white on transparent.20160208
  • About
  • Editors
  • Consulting Editors
  • For authors
  • Alerts
  • Advertising/recruitment
  • Subscribe
  • Contact
  • Current Issue
  • Past Issues
  • By specialty
    • Cardiology
    • Gastroenterology
    • Immunology
    • Metabolism
    • Nephrology
    • Neuroscience
    • Oncology
    • Pulmonology
    • Vascular biology
    • All...
  • Videos
    • Conversations with Giants in Medicine
    • Author's Takes
  • Reviews
    • View all reviews...
    • Cellular senescence in human disease (Apr 2018)
    • Fibrosis (Jan 2018)
    • Glia and Neurodegeneration (Sep 2017)
    • Transplantation (Jun 2017)
    • Nuclear Receptors (Apr 2017)
    • Metabolism and Inflammation (Jan 2017)
    • Hypoxia and Inflammation (Oct 2016)
    • View all review series...
  • Collections
    • Recently published
    • Commentaries
    • Concise Communication
    • Editorials
    • Opinion
    • Scientific Show Stoppers
    • Top read articles
    • In-Press Preview
  • Clinical Medicine
  • JCI This Month
    • Current issue
    • Past issues

Jci only white

  • About
  • Editors
  • Consulting Editors
  • For authors
  • Current issue
  • Past issues
  • By specialty
  • Subscribe
  • Alerts
  • Advertise
  • Contact
  • Conversations with Giants in Medicine
  • Author's Takes
  • Recently published
  • Brief Reports
  • Technical Advances
  • Commentaries
  • Editorials
  • Hindsight
  • Review series
  • Reviews
  • The Attending Physician
  • First Author Perspectives
  • Scientific Show Stoppers
  • Top read articles
  • Concise Communication

Top read articles in the last 30 days

This list is updated daily and reflects the last month of access data. Articles older than two years will not be shown.

  • Research
  • Review
Targeting of nonlipidated, aggregated apoE with antibodies inhibits amyloid accumulation
Fan Liao, … , Ryan J. Watts, David M. Holtzman
Fan Liao, … , Ryan J. Watts, David M. Holtzman
Published March 30, 2018
Citation Information: J Clin Invest. 2018. https://doi.org/10.1172/JCI96429.
View: Text | PDF
Categories: Research Article Neuroscience

Targeting of nonlipidated, aggregated apoE with antibodies inhibits amyloid accumulation

  • Text
  • PDF
Abstract

The apolipoprotein E E4 allele of the APOE gene is the strongest genetic factor for late-onset Alzheimer disease (LOAD). There is compelling evidence that apoE influences Alzheimer disease (AD) in large part by affecting amyloid β (Aβ) aggregation and clearance; however, the molecular mechanism underlying these findings remains largely unknown. Herein, we tested whether anti–human apoE antibodies can decrease Aβ pathology in mice producing both human Aβ and apoE4, and investigated the mechanism underlying these effects. We utilized APPPS1-21 mice crossed to apoE4-knockin mice expressing human apoE4 (APPPS1-21/APOE4). We discovered an anti–human apoE antibody, anti–human apoE 4 (HAE-4), that specifically recognizes human apoE4 and apoE3 and preferentially binds nonlipidated, aggregated apoE over the lipidated apoE found in circulation. HAE-4 also binds to apoE in amyloid plaques in unfixed brain sections and in living APPPS1-21/APOE4 mice. When delivered centrally or by peripheral injection, HAE-4 reduced Aβ deposition in APPPS1-21/APOE4 mice. Using adeno-associated virus to express 2 different full-length anti–apoE antibodies in the brain, we found that HAE antibodies decreased amyloid accumulation, which was dependent on Fcγ receptor function. These data support the hypothesis that a primary mechanism for apoE-mediated plaque formation may be a result of apoE aggregation, as preferentially targeting apoE aggregates with therapeutic antibodies reduces Aβ pathology and may represent a selective approach to treat AD.

Authors

Fan Liao, Aimin Li, Monica Xiong, Nga Bien-Ly, Hong Jiang, Yin Zhang, Mary Beth Finn, Rosa Hoyle, Jennifer Keyser, Katheryn B. Lefton, Grace O. Robinson, Javier Remolina Serrano, Adam P. Silverman, Jing L. Guo, Jennifer Getz, Kirk Henne, Cheryl E.G. Leyns, Gilbert Gallardo, Jason D. Ulrich, Patrick M. Sullivan, Eli Paul Lerner, Eloise Hudry, Zachary K. Sweeney, Mark S. Dennis, Bradley T. Hyman, Ryan J. Watts, David M. Holtzman

×

Total views: 3293


Insulin resistance causes inflammation in adipose tissue
Mitsugu Shimobayashi, … , Ralph Peterli, Michael N. Hall
Mitsugu Shimobayashi, … , Ralph Peterli, Michael N. Hall
Published April 2, 2018
Citation Information: J Clin Invest. 2018;128(4):1538-1550. https://doi.org/10.1172/JCI96139.
View: Text | PDF
Categories: Research Article Inflammation Metabolism

Insulin resistance causes inflammation in adipose tissue

  • Text
  • PDF
Abstract

Obesity is a major risk factor for insulin resistance and type 2 diabetes. In adipose tissue, obesity-mediated insulin resistance correlates with the accumulation of proinflammatory macrophages and inflammation. However, the causal relationship of these events is unclear. Here, we report that obesity-induced insulin resistance in mice precedes macrophage accumulation and inflammation in adipose tissue. Using a mouse model that combines genetically induced, adipose-specific insulin resistance (mTORC2-knockout) and diet-induced obesity, we found that insulin resistance causes local accumulation of proinflammatory macrophages. Mechanistically, insulin resistance in adipocytes results in production of the chemokine monocyte chemoattractant protein 1 (MCP1), which recruits monocytes and activates proinflammatory macrophages. Finally, insulin resistance (high homeostatic model assessment of insulin resistance [HOMA-IR]) correlated with reduced insulin/mTORC2 signaling and elevated MCP1 production in visceral adipose tissue from obese human subjects. Our findings suggest that insulin resistance in adipose tissue leads to inflammation rather than vice versa.

Authors

Mitsugu Shimobayashi, Verena Albert, Bettina Woelnerhanssen, Irina C. Frei, Diana Weissenberger, Anne Christin Meyer-Gerspach, Nicolas Clement, Suzette Moes, Marco Colombi, Jerome A. Meier, Marta M. Swierczynska, Paul Jenö, Christoph Beglinger, Ralph Peterli, Michael N. Hall

×

Total views: 2991


JAM3 maintains leukemia-initiating cell self-renewal through LRP5/AKT/β-catenin/CCND1 signaling
Yaping Zhang, … , Guo-Qiang Chen, Junke Zheng
Yaping Zhang, … , Guo-Qiang Chen, Junke Zheng
Published March 26, 2018
Citation Information: J Clin Invest. 2018. https://doi.org/10.1172/JCI93198.
View: Text | PDF
Categories: Research Article Hematology Stem cells

JAM3 maintains leukemia-initiating cell self-renewal through LRP5/AKT/β-catenin/CCND1 signaling

  • Text
  • PDF
Abstract

Leukemia-initiating cells (LICs) are responsible for the initiation, development, and relapse of leukemia. The identification of novel therapeutic LIC targets is critical to curing leukemia. In this report, we reveal that junctional adhesion molecule 3 (JAM3) is highly enriched in both mouse and human LICs. Leukemogenesis is almost completely abrogated upon Jam3 deletion during serial transplantations in an MLL-AF9–induced murine acute myeloid leukemia model. In contrast, Jam3 deletion does not affect the functions of mouse hematopoietic stem cells. Moreover, knockdown of JAM3 leads to a dramatic decrease in the proliferation of both human leukemia cell lines and primary LICs. JAM3 directly associates with LRP5 to activate the downstream PDK1/AKT pathway, followed by the downregulation of GSK3β and activation of β-catenin/CCND1 signaling, to maintain the self-renewal ability and cell cycle entry of LICs. Thus, JAM3 may serve as a functional LIC marker and play an important role in the maintenance of LIC stemness through unexpected LRP5/PDK1/AKT/GSK3β/β-catenin/CCND1 signaling pathways but not via its canonical role in cell junctions and migration. JAM3 may be an ideal therapeutic target for the eradication of LICs without influencing normal hematopoiesis.

Authors

Yaping Zhang, Fangzhen Xia, Xiaoye Liu, Zhuo Yu, Li Xie, Ligen Liu, Chiqi Chen, Haishan Jiang, Xiaoxin Hao, Xiaoxiao He, Feifei Zhang, Hao Gu, Jun Zhu, Haitao Bai, Cheng Cheng Zhang, Guo-Qiang Chen, Junke Zheng

×

Total views: 2942


Blocking fatty acid–fueled mROS production within macrophages alleviates acute gouty inflammation
Christopher J. Hall, … , Nicola Dalbeth, Philip S. Crosier
Christopher J. Hall, … , Nicola Dalbeth, Philip S. Crosier
Published March 26, 2018
Citation Information: J Clin Invest. 2018. https://doi.org/10.1172/JCI94584.
View: Text | PDF
Categories: Research Article Immunology Inflammation

Blocking fatty acid–fueled mROS production within macrophages alleviates acute gouty inflammation

  • Text
  • PDF
Abstract

Gout is the most common inflammatory arthritis affecting men. Acute gouty inflammation is triggered by monosodium urate (MSU) crystal deposition in and around joints that activates macrophages into a proinflammatory state, resulting in neutrophil recruitment. A complete understanding of how MSU crystals activate macrophages in vivo has been difficult because of limitations of live imaging this process in traditional animal models. By live imaging the macrophage and neutrophil response to MSU crystals within an intact host (larval zebrafish), we reveal that macrophage activation requires mitochondrial ROS (mROS) generated through fatty acid oxidation. This mitochondrial source of ROS contributes to NF-κB–driven production of IL-1β and TNF-α, which promote neutrophil recruitment. We demonstrate the therapeutic utility of this discovery by showing that this mechanism is conserved in human macrophages and, via pharmacologic blockade, that it contributes to neutrophil recruitment in a mouse model of acute gouty inflammation. To our knowledge, this study is the first to uncover an immunometabolic mechanism of macrophage activation that operates during acute gouty inflammation. Targeting this pathway holds promise in the management of gout and, potentially, other macrophage-driven diseases.

Authors

Christopher J. Hall, Leslie E. Sanderson, Lisa M. Lawrence, Bregina Pool, Maarten van der Kroef, Elina Ashimbayeva, Denver Britto, Jacquie L. Harper, Graham J. Lieschke, Jonathan W. Astin, Kathryn E. Crosier, Nicola Dalbeth, Philip S. Crosier

×

Total views: 2573


Imaging activated T cells predicts response to cancer vaccines
Israt S. Alam, … , Ronald Levy, Sanjiv S. Gambhir
Israt S. Alam, … , Ronald Levy, Sanjiv S. Gambhir
Published March 29, 2018
Citation Information: J Clin Invest. 2018. https://doi.org/10.1172/JCI98509.
View: Text | PDF
Categories: In-Press Preview Research Article Immunology Oncology

Imaging activated T cells predicts response to cancer vaccines

  • Text
  • PDF
Abstract

In situ cancer vaccines are under active clinical investigation due to their reported ability to eradicate both local and disseminated malignancies. Intratumoral vaccine administration is thought to activate a T cell mediated immune response, which begins in the treated tumor and cascades systemically. We describe a positron emission tomography tracer (64Cu-DOTA-AbOX40) that enabled non-invasive and longitudinal imaging of OX40, a cell surface marker of T cell activation. We report the spatiotemporal dynamics of T cell activation following in situ vaccination with CpG oligodeoxynucleotide, in a dual tumor bearing mouse model. We demonstrate that OX40 imaging could predict tumor responses at day 9 post treatment based on tumor tracer uptake at day 2, with higher accuracy than both anatomical and blood-based measurements. These studies provide key insights into global T cell activation following local CpG treatment and indicate that 64Cu-DOTA-AbOX40 is a promising candidate for monitoring clinical cancer immunotherapy strategies.

Authors

Israt S. Alam, Aaron T. Mayer, Idit Sagiv-Barfi, Kezheng Wang, Ophir Vermesh, Debra K. Czerwinski, Emily M. Johnson, Michelle L. James, Ronald Levy, Sanjiv S. Gambhir

×

Total views: 2554


PD-L1 in tumor microenvironment mediates resistance to oncolytic immunotherapy
Dmitriy Zamarin, … , Taha Merghoub, Jedd D. Wolchok
Dmitriy Zamarin, … , Taha Merghoub, Jedd D. Wolchok
Published April 2, 2018
Citation Information: J Clin Invest. 2018;128(4):1413-1428. https://doi.org/10.1172/JCI98047.
View: Text | PDF
Categories: Research Article Immunology Oncology

PD-L1 in tumor microenvironment mediates resistance to oncolytic immunotherapy

  • Text
  • PDF
Abstract

Intralesional therapy with oncolytic viruses (OVs) leads to the activation of local and systemic immune pathways, which may present targets for further combinatorial therapies. Here, we used human tumor histocultures as well as syngeneic tumor models treated with Newcastle disease virus (NDV) to identify a range of immune targets upregulated with OV treatment. Despite tumor infiltration of effector T lymphocytes in response to NDV, there was ongoing inhibition through programmed death ligand 1 (PD-L1), acting as a mechanism of early and late adaptive immune resistance to the type I IFN response and T cell infiltration, respectively. Systemic therapeutic targeting of programmed cell death receptor 1 (PD-1) or PD-L1 in combination with intratumoral NDV resulted in the rejection of both treated and distant tumors. These findings have implications for the timing of PD-1/PD-L1 blockade in conjunction with OV therapy and highlight the importance of understanding the adaptive mechanisms of immune resistance to specific OVs for the rational design of combinatorial approaches using these agents.

Authors

Dmitriy Zamarin, Jacob M. Ricca, Svetlana Sadekova, Anton Oseledchyk, Ying Yu, Wendy M. Blumenschein, Jerelyn Wong, Mathieu Gigoux, Taha Merghoub, Jedd D. Wolchok

×

Total views: 2452


Peptide mimic for influenza vaccination using nonnatural combinatorial chemistry
John J. Miles, … , David A. Price, Andrew K. Sewell
John J. Miles, … , David A. Price, Andrew K. Sewell
Published April 2, 2018
Citation Information: J Clin Invest. 2018;128(4):1569-1580. https://doi.org/10.1172/JCI91512.
View: Text | PDF
Categories: Research Article Immunology Infectious disease

Peptide mimic for influenza vaccination using nonnatural combinatorial chemistry

  • Text
  • PDF
Abstract

Polypeptide vaccines effectively activate human T cells but suffer from poor biological stability, which confines both transport logistics and in vivo therapeutic activity. Synthetic biology has the potential to address these limitations through the generation of highly stable antigenic “mimics” using subunits that do not exist in the natural world. We developed a platform based on D–amino acid combinatorial chemistry and used this platform to reverse engineer a fully artificial CD8+ T cell agonist that mirrored the immunogenicity profile of a native epitope blueprint from influenza virus. This nonnatural peptide was highly stable in human serum and gastric acid, reflecting an intrinsic resistance to physical and enzymatic degradation. In vitro, the synthetic agonist stimulated and expanded an archetypal repertoire of polyfunctional human influenza virus–specific CD8+ T cells. In vivo, specific responses were elicited in naive humanized mice by subcutaneous vaccination, conferring protection from subsequent lethal influenza challenge. Moreover, the synthetic agonist was immunogenic after oral administration. This proof-of-concept study highlights the power of synthetic biology to expand the horizons of vaccine design and therapeutic delivery.

Authors

John J. Miles, Mai Ping Tan, Garry Dolton, Emily S.J. Edwards, Sarah A.E. Galloway, Bruno Laugel, Mathew Clement, Julia Makinde, Kristin Ladell, Katherine K. Matthews, Thomas S. Watkins, Katie Tungatt, Yide Wong, Han Siean Lee, Richard J. Clark, Johanne M. Pentier, Meriem Attaf, Anya Lissina, Ann Ager, Awen Gallimore, Pierre J. Rizkallah, Stephanie Gras, Jamie Rossjohn, Scott R. Burrows, David K. Cole, David A. Price, Andrew K. Sewell

×

Total views: 2399


Autoinhibitory regulation of S100A8/S100A9 alarmin activity locally restricts sterile inflammation
Thomas Vogl, … , Thomas Pap, Johannes Roth
Thomas Vogl, … , Thomas Pap, Johannes Roth
Published April 3, 2018
Citation Information: J Clin Invest. 2018. https://doi.org/10.1172/JCI89867.
View: Text | PDF
Categories: Research Article Autoimmunity Inflammation

Autoinhibitory regulation of S100A8/S100A9 alarmin activity locally restricts sterile inflammation

  • Text
  • PDF
Abstract

Autoimmune diseases, such as psoriasis and arthritis, show a patchy distribution of inflammation despite systemic dysregulation of adaptive immunity. Thus, additional tissue-derived signals, such as danger-associated molecular patterns (DAMPs), are indispensable for manifestation of local inflammation. S100A8/S100A9 complexes are the most abundant DAMPs in many autoimmune diseases. However, regulatory mechanisms locally restricting DAMP activities are barely understood. We now unravel for the first time, to our knowledge, a mechanism of autoinhibition in mice and humans restricting S100-DAMP activity to local sites of inflammation. Combining protease degradation, pull-down assays, mass spectrometry, and targeted mutations, we identified specific peptide sequences within the second calcium-binding EF-hands triggering TLR4/MD2-dependent inflammation. These binding sites are free when S100A8/S100A9 heterodimers are released at sites of inflammation. Subsequently, S100A8/S100A9 activities are locally restricted by calcium-induced (S100A8/S100A9)2 tetramer formation hiding the TLR4/MD2-binding site within the tetramer interphase, thus preventing undesirable systemic effects. Loss of this autoinhibitory mechanism in vivo results in TNF-α–driven fatal inflammation, as shown by lack of tetramer formation in crossing S100A9–/– mice with 2 independent TNF-α–transgene mouse strains. Since S100A8/S100A9 is the most abundant DAMP in many inflammatory diseases, specifically blocking the TLR4-binding site of active S100 dimers may represent a promising approach for local suppression of inflammatory diseases, avoiding systemic side effects.

Authors

Thomas Vogl, Athanasios Stratis, Viktor Wixler, Tom Völler, Sumita Thurainayagam, Selina K. Jorch, Stefanie Zenker, Alena Dreiling, Deblina Chakraborty, Mareike Fröhling, Peter Paruzel, Corinna Wehmeyer, Sven Hermann, Olympia Papantonopoulou, Christiane Geyer, Karin Loser, Michael Schäfers, Stephan Ludwig, Monika Stoll, Tomas Leanderson, Joachim L. Schultze, Simone König, Thomas Pap, Johannes Roth

×

Total views: 2240


TNF-driven adaptive response mediates resistance to EGFR inhibition in lung cancer
Ke Gong, … , Dawen Zhao, Amyn A. Habib
Ke Gong, … , Dawen Zhao, Amyn A. Habib
Published April 3, 2018
Citation Information: J Clin Invest. 2018. https://doi.org/10.1172/JCI96148.
View: Text | PDF
Categories: In-Press Preview Research Article Therapeutics

TNF-driven adaptive response mediates resistance to EGFR inhibition in lung cancer

  • Text
  • PDF
Abstract

Although aberrant Epidermal Growth Factor Receptor (EGFR) signaling is widespread in cancer, EGFR inhibition is effective only in a subset of NSCLC (non-small cell lung cancer) with EGFR activating mutations. A majority of NSCLCs express EGFR wild type (EGFRwt) and do not respond to EGFR inhibition. Tumor necrosis factor (TNF) is a major mediator of inflammation-induced cancer. We find that a rapid increase in TNF level is a universal adaptive response to EGFR inhibition in NSCLC regardless of EGFR status. EGFR signaling actively suppresses TNF mRNA levels by inducing expression of miR-21 resulting in decreased TNF mRNA stability. Conversely, EGFR inhibition results in loss of miR-21 and increased TNF mRNA stability. In addition, TNF-induced NF-kB activation leads to increased TNF transcription in a feedforward loop. Inhibition of TNF signaling renders EGFRwt expressing NSCLC cell lines and an EGFRwt Patient-Derived Xenograft (PDX) model highly sensitive to EGFR inhibition. In EGFR mutant oncogene-addicted cells, blocking TNF enhances the effectiveness of EGFR inhibition. EGFR plus TNF inhibition is also effective in NSCLC with acquired resistance to EGFR inhibition. We suggest concomitant EGFR and TNF inhibition as a new treatment approach that could be beneficial for a majority of lung cancer patients.

Authors

Ke Gong, Gao Guo, David E. Gerber, Boning Gao, Michael Peyton, Chun Huang, John D. Minna, Kimmo J. Hatanpaa, Kemp Kernstine, Ling Cai, Yang Xie, Hong Zhu, Farjana Fattah, Shanrong Zhang, Masaya Takahashi, Bipasha Mukherjee, Sandeep Burma, Jonathan Dowell, Kathryn Dao, Vassiliki A. Papadimitrakopoulou, Victor Olivas, Trever G. Bivona, Dawen Zhao, Amyn A. Habib

×

Total views: 2218


PAI1 mediates fibroblast–mast cell interactions in skin fibrosis
Neha Pincha, … , Paul Mazhuvanchary Jacob, Colin Jamora
Neha Pincha, … , Paul Mazhuvanchary Jacob, Colin Jamora
Published March 26, 2018
Citation Information: J Clin Invest. 2018. https://doi.org/10.1172/JCI99088.
View: Text | PDF
Categories: Research Article Cell biology Inflammation

PAI1 mediates fibroblast–mast cell interactions in skin fibrosis

  • Text
  • PDF
Abstract

Fibrosis is a prevalent pathological condition arising from the chronic activation of fibroblasts. This activation results from the extensive intercellular crosstalk mediated by both soluble factors and direct cell-cell connections. Prominent among these are the interactions of fibroblasts with immune cells, in which the fibroblast–mast cell connection, although acknowledged, is relatively unexplored. We have used a Tg mouse model of skin fibrosis, based on expression of the transcription factor Snail in the epidermis, to probe the mechanisms regulating mast cell activity and the contribution of these cells to this pathology. We have discovered that Snail-expressing keratinocytes secrete plasminogen activator inhibitor type 1 (PAI1), which functions as a chemotactic factor to increase mast cell infiltration into the skin. Moreover, we have determined that PAI1 upregulates intercellular adhesion molecule type 1 (ICAM1) expression on dermal fibroblasts, rendering them competent to bind to mast cells. This heterotypic cell-cell adhesion, also observed in the skin fibrotic disorder scleroderma, culminates in the reciprocal activation of both mast cells and fibroblasts, leading to the cascade of events that promote fibrogenesis. Thus, we have identified roles for PAI1 in the multifactorial program of fibrogenesis that expand its functional repertoire beyond its canonical role in plasmin-dependent processes.

Authors

Neha Pincha, Edries Yousaf Hajam, Krithika Badarinath, Surya Prakash Rao Batta, Tafheem Masudi, Rakesh Dey, Peter Andreasen, Toshiaki Kawakami, Rekha Samuel, Renu George, Debashish Danda, Paul Mazhuvanchary Jacob, Colin Jamora

×

Total views: 2134

Show more results Load more

Mechanisms and functions of cellular senescence
Nicolás Herranz, Jesús Gil
Nicolás Herranz, Jesús Gil
Published April 2, 2018
Citation Information: J Clin Invest. 2018;128(4):1238-1246. https://doi.org/10.1172/JCI95148.
View: Text | PDF
Category: Review Series

Mechanisms and functions of cellular senescence

  • Text
  • PDF
Abstract

Cellular senescence is a highly stable cell cycle arrest that is elicited in response to different stresses. By imposing a growth arrest, senescence limits the replication of old or damaged cells. Besides exiting the cell cycle, senescent cells undergo many other phenotypic alterations such as metabolic reprogramming, chromatin rearrangement, or autophagy modulation. In addition, senescent cells produce and secrete a complex combination of factors, collectively referred as the senescence-associated secretory phenotype, that mediate most of their non–cell-autonomous effects. Because senescent cells influence the outcome of a variety of physiological and pathological processes, including cancer and age-related diseases, pro-senescent and anti-senescent therapies are actively being explored. In this Review, we discuss the mechanisms regulating different aspects of the senescence phenotype and their functional implications. This knowledge is essential to improve the identification and characterization of senescent cells in vivo and will help to develop rational strategies to modulate the senescence program for therapeutic benefit.

Authors

Nicolás Herranz, Jesús Gil

×

Total views: 2642


Senescent cells: a therapeutic target for cardiovascular disease
Bennett G. Childs, … , Hu Li, Jan M. van Deursen
Bennett G. Childs, … , Hu Li, Jan M. van Deursen
Published April 2, 2018
Citation Information: J Clin Invest. 2018;128(4):1217-1228. https://doi.org/10.1172/JCI95146.
View: Text | PDF
Category: Review Series

Senescent cells: a therapeutic target for cardiovascular disease

  • Text
  • PDF
Abstract

Cellular senescence, a major tumor-suppressive cell fate, has emerged from humble beginnings as an in vitro phenomenon into recognition as a fundamental mechanism of aging. In the process, senescent cells have attracted attention as a therapeutic target for age-related diseases, including cardiovascular disease (CVD), the leading cause of morbidity and mortality in the elderly. Given the aging global population and the inadequacy of current medical management, attenuating the health care burden of CVD would be transformative to clinical practice. Here, we review the evidence that cellular senescence drives CVD in a bimodal fashion by both priming the aged cardiovascular system for disease and driving established disease forward. Hence, the growing field of senotherapy (neutralizing senescent cells for therapeutic benefit) is poised to contribute to both prevention and treatment of CVD.

Authors

Bennett G. Childs, Hu Li, Jan M. van Deursen

×

Total views: 2415


Strategies targeting cellular senescence
Yossi Ovadya, Valery Krizhanovsky
Yossi Ovadya, Valery Krizhanovsky
Published April 2, 2018
Citation Information: J Clin Invest. 2018;128(4):1247-1254. https://doi.org/10.1172/JCI95149.
View: Text | PDF
Category: Review Series

Strategies targeting cellular senescence

  • Text
  • PDF
Abstract

Cellular senescence is a physiological phenomenon that has both beneficial and detrimental consequences. Senescence limits tumorigenesis and tissue damage throughout the lifetime. However, at the late stages of life, senescent cells increasingly accumulate in tissues and might also contribute to the development of various age-related pathologies. Recent studies have revealed the molecular pathways that preserve the viability of senescent cells and the ones regulating their immune surveillance. These studies provide essential initial insights for the development of novel therapeutic strategies for targeting senescent cells. At the same time they stress the need to understand the limitations of the existing strategies, their efficacy and safety, and the possible deleterious consequences of senescent cell elimination. Here we discuss the existing strategies for targeting senescent cells and upcoming challenges in translating these strategies into safe and efficient therapies. Successful translation of these strategies could have implications for treating a variety of diseases at old age and could potentially reshape our view of health management during aging.

Authors

Yossi Ovadya, Valery Krizhanovsky

×

Total views: 2334


Senescent cells and osteoarthritis: a painful connection
Ok Hee Jeon, … , Judith Campisi, Jennifer H. Elisseeff
Ok Hee Jeon, … , Judith Campisi, Jennifer H. Elisseeff
Published April 2, 2018
Citation Information: J Clin Invest. 2018;128(4):1229-1237. https://doi.org/10.1172/JCI95147.
View: Text | PDF
Category: Review Series

Senescent cells and osteoarthritis: a painful connection

  • Text
  • PDF
Abstract

Senescent cells (SnCs) are associated with age-related pathologies. Osteoarthritis is a chronic disease characterized by pain, loss of cartilage, and joint inflammation, and its incidence increases with age. For years, the presence of SnCs in cartilage isolated from patients undergoing total knee artificial implants has been noted, but these cells’ relevance to disease was unclear. In this Review, we summarize current knowledge of SnCs in the multiple tissues that constitute the articular joint. New evidence for the causative role of SnCs in the development of posttraumatic and age-related arthritis is reviewed along with the therapeutic benefit of SnC clearance. As part of their senescence-associated secretory phenotype, SnCs secrete cytokines that impact the immune system and its response to joint tissue trauma. We present concepts of the immune response to tissue trauma as well as the interactions with SnCs and the local tissue environment. Finally, we discuss therapeutic implications of targeting SnCs in treating osteoarthritis.

Authors

Ok Hee Jeon, Nathaniel David, Judith Campisi, Jennifer H. Elisseeff

×

Total views: 2005


Cellular senescence in brain aging and neurodegenerative diseases: evidence and perspectives
Darren J. Baker, Ronald C. Petersen
Darren J. Baker, Ronald C. Petersen
Published April 2, 2018
Citation Information: J Clin Invest. 2018;128(4):1208-1216. https://doi.org/10.1172/JCI95145.
View: Text | PDF
Category: Review Series

Cellular senescence in brain aging and neurodegenerative diseases: evidence and perspectives

  • Text
  • PDF
Abstract

Along with a general decline in overall health, most chronic degenerative human diseases are inherently associated with increasing age. Age-associated cognitive impairments and neurodegenerative diseases, such as Parkinson’s and Alzheimer’s diseases, are potentially debilitating conditions that lack viable options for treatment, resulting in a tremendous economic and societal cost. Most high-profile clinical trials for neurodegenerative diseases have led to inefficacious results, suggesting that novel approaches to treating these pathologies are needed. Numerous recent studies have demonstrated that senescent cells, which are characterized by sustained cell cycle arrest and production of a distinct senescence-associated secretory phenotype, accumulate with age and at sites of age-related diseases throughout the body, where they actively promote tissue deterioration. Cells with features of senescence have been detected in the context of brain aging and neurodegenerative disease, suggesting that they may also promote dysfunction. Here, we discuss the evidence implicating senescent cells in neurodegenerative diseases, the mechanistic contribution of these cells that may actively drive neurodegeneration, and how these cells or their effects may be targeted therapeutically.

Authors

Darren J. Baker, Ronald C. Petersen

×

Total views: 1237


Fructose metabolism and metabolic disease
Sarah A. Hannou, … , Nicola M. McKeown, Mark A. Herman
Sarah A. Hannou, … , Nicola M. McKeown, Mark A. Herman
Published February 1, 2018
Citation Information: J Clin Invest. 2018;128(2):545-555. https://doi.org/10.1172/JCI96702.
View: Text | PDF
Category: Review

Fructose metabolism and metabolic disease

  • Text
  • PDF
Abstract

Increased sugar consumption is increasingly considered to be a contributor to the worldwide epidemics of obesity and diabetes and their associated cardiometabolic risks. As a result of its unique metabolic properties, the fructose component of sugar may be particularly harmful. Diets high in fructose can rapidly produce all of the key features of the metabolic syndrome. Here we review the biology of fructose metabolism as well as potential mechanisms by which excessive fructose consumption may contribute to cardiometabolic disease.

Authors

Sarah A. Hannou, Danielle E. Haslam, Nicola M. McKeown, Mark A. Herman

×

Total views: 934


Microglia in Alzheimer’s disease
Heela Sarlus, Michael T. Heneka
Heela Sarlus, Michael T. Heneka
Published September 1, 2017
Citation Information: J Clin Invest. 2017;127(9):3240-3249. https://doi.org/10.1172/JCI90606.
View: Text | PDF
Category: Review Series

Microglia in Alzheimer’s disease

  • Text
  • PDF
Abstract

Microglia are brain-resident myeloid cells that mediate key functions to support the CNS. Microglia express a wide range of receptors that act as molecular sensors, which recognize exogenous or endogenous CNS insults and initiate an immune response. In addition to their classical immune cell function, microglia act as guardians of the brain by promoting phagocytic clearance and providing trophic support to ensure tissue repair and maintain cerebral homeostasis. Conditions associated with loss of homeostasis or tissue changes induce several dynamic microglial processes, including changes of cellular morphology, surface phenotype, secretory mediators, and proliferative responses (referred to as an “activated state”). Activated microglia represent a common pathological feature of several neurodegenerative diseases, including Alzheimer’s disease (AD). Cumulative evidence suggests that microglial inflammatory activity in AD is increased while microglial-mediated clearance mechanisms are compromised. Microglia are perpetually engaged in a mutual interaction with the surrounding environment in CNS; thus, diverse microglial reactions at different disease stages may open new avenues for therapeutic intervention and modification of inflammatory activities. In this Review, the role of microglia in the pathogenesis of AD and the modulation of microglia activity as a therapeutic modality will be discussed.

Authors

Heela Sarlus, Michael T. Heneka

×

Total views: 711


Inflammatory mechanisms linking obesity and metabolic disease
Alan R. Saltiel, Jerrold M. Olefsky
Alan R. Saltiel, Jerrold M. Olefsky
Published January 3, 2017
Citation Information: J Clin Invest. 2017;127(1):1-4. https://doi.org/10.1172/JCI92035.
View: Text | PDF
Category: Review Series

Inflammatory mechanisms linking obesity and metabolic disease

  • Text
  • PDF
Abstract

There are currently over 1.9 billion people who are obese or overweight, leading to a rise in related health complications, including insulin resistance, type 2 diabetes, cardiovascular disease, liver disease, cancer, and neurodegeneration. The finding that obesity and metabolic disorder are accompanied by chronic low-grade inflammation has fundamentally changed our view of the underlying causes and progression of obesity and metabolic syndrome. We now know that an inflammatory program is activated early in adipose expansion and during chronic obesity, permanently skewing the immune system to a proinflammatory phenotype, and we are beginning to delineate the reciprocal influence of obesity and inflammation. Reviews in this series examine the activation of the innate and adaptive immune system in obesity; inflammation within diabetic islets, brain, liver, gut, and muscle; the role of inflammation in fibrosis and angiogenesis; the factors that contribute to the initiation of inflammation; and therapeutic approaches to modulate inflammation in the context of obesity and metabolic syndrome.

Authors

Alan R. Saltiel, Jerrold M. Olefsky

×

Total views: 481


Understanding the functions and relationships of the glymphatic system and meningeal lymphatics
Antoine Louveau, … , Maiken Nedergaard, Jonathan Kipnis
Antoine Louveau, … , Maiken Nedergaard, Jonathan Kipnis
Published September 1, 2017
Citation Information: J Clin Invest. 2017;127(9):3210-3219. https://doi.org/10.1172/JCI90603.
View: Text | PDF
Category: Review Series

Understanding the functions and relationships of the glymphatic system and meningeal lymphatics

  • Text
  • PDF
Abstract

Recent discoveries of the glymphatic system and of meningeal lymphatic vessels have generated a lot of excitement, along with some degree of skepticism. Here, we summarize the state of the field and point out the gaps of knowledge that should be filled through further research. We discuss the glymphatic system as a system that allows CNS perfusion by the cerebrospinal fluid (CSF) and interstitial fluid (ISF). We also describe the recently characterized meningeal lymphatic vessels and their role in drainage of the brain ISF, CSF, CNS-derived molecules, and immune cells from the CNS and meninges to the peripheral (CNS-draining) lymph nodes. We speculate on the relationship between the two systems and their malfunction that may underlie some neurological diseases. Although much remains to be investigated, these new discoveries have changed our understanding of mechanisms underlying CNS immune privilege and CNS drainage. Future studies should explore the communications between the glymphatic system and meningeal lymphatics in CNS disorders and develop new therapeutic modalities targeting these systems.

Authors

Antoine Louveau, Benjamin A. Plog, Salli Antila, Kari Alitalo, Maiken Nedergaard, Jonathan Kipnis

×

Total views: 474


Blood will out: vascular contributions to Alzheimer’s disease
Sidney Strickland
Sidney Strickland
Published February 1, 2018
Citation Information: J Clin Invest. 2018;128(2):556-563. https://doi.org/10.1172/JCI97509.
View: Text | PDF
Category: Review

Blood will out: vascular contributions to Alzheimer’s disease

  • Text
  • PDF
Abstract

The fundamental pathology in Alzheimer’s disease (AD) is neuronal dysfunction leading to cognitive impairment. The amyloid-β peptide (Aβ), derived from amyloid precursor protein, is one driver of AD, but how it leads to neuronal dysfunction is not established. In this Review, I discuss the complexity of AD and possible cause-and-effect relationships between Aβ and the vascular and hemostatic systems. AD can be considered a multifactorial syndrome with various contributing pathological mechanisms. Therefore, as is routinely done with cancer, it will be important to classify patients with respect to their disease signature so that specific pathologies, including vascular pathways, can be therapeutically targeted.

Authors

Sidney Strickland

×

Total views: 426

Show more results Load more

Advertisement
Follow JCI: Facebook logo white Twitter logo v2 Rss icon
Copyright © 2018 American Society for Clinical Investigation
ISSN: 0021-9738 (print), 1558-8238 (online)

Sign up for email alerts