1
|
Mehta SK, Diak DM, Bustos-Lopez S, Nelman-Gonzalez M, Chen X, Plante I, Stray SJ, Tandon R, Crucian BE. Effect of Simulated Cosmic Radiation on Cytomegalovirus Reactivation and Lytic Replication. Int J Mol Sci 2024; 25:10337. [PMID: 39408667 PMCID: PMC11477029 DOI: 10.3390/ijms251910337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 09/19/2024] [Accepted: 09/21/2024] [Indexed: 10/20/2024] Open
Abstract
Human exploration of the solar system will expose crew members to galactic cosmic radiation (GCR), with a potential for adverse health effects. GCR particles (protons and ions) move at nearly the speed of light and easily penetrate space station walls, as well as the human body. Previously, we have shown reactivation of latent herpesviruses, including herpes simplex virus, Varicella zoster virus, Epstein-Barr virus, and cytomegalovirus (CMV), during stays at the International Space Station. Given the prevalence of latent CMV and the known propensity of space radiation to cause alterations in many cellular processes, we undertook this study to understand the role of GCR in reactivating latent CMV. Latently infected Kasumi cells with CMV were irradiated with 137Cs gamma rays, 150 MeV protons, 600 MeV/n carbon ions, 600 MeV/n iron ions, proton ions, and simulated GCR. The CMV copy number increased significantly in the cells exposed to radiation as compared with the non-irradiated controls. Viral genome sequencing did not reveal significant nucleotide differences among the compared groups. However, transcriptome analysis showed the upregulation of transcription of the UL49 ORF, implicating it in the switch from latent to lytic replication. These findings support our hypothesis that GCR may be a strong contributor to the reactivation of CMV infection seen in ISS crew members.
Collapse
Affiliation(s)
- Satish K. Mehta
- JES Tech, NASA, Johnson Space Center, Houston, TX 77058, USA
| | | | - Sara Bustos-Lopez
- Department of Health and Human Performance, University of Houston, Houston, TX 77004, USA;
| | | | - Xi Chen
- KBR, Houston, TX 77002, USA; (M.N.-G.); (X.C.); (I.P.)
| | - Ianik Plante
- KBR, Houston, TX 77002, USA; (M.N.-G.); (X.C.); (I.P.)
| | - Stephen J. Stray
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS 39216, USA; (S.J.S.); (R.T.)
| | - Ritesh Tandon
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS 39216, USA; (S.J.S.); (R.T.)
| | | |
Collapse
|
2
|
Sukhina A, Queriault C, Hall E, Rome K, Aggarwal M, Nunn E, Weiss A, Nguyen J, Bailis W. Malnutrition drives infection susceptibility and dysregulated myelopoiesis that persists after refeeding intervention. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.19.608575. [PMID: 39229137 PMCID: PMC11370435 DOI: 10.1101/2024.08.19.608575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Undernutrition is one of the largest persistent global health crises, with nearly 1 billion people facing severe food insecurity. Infectious disease represents the main underlying cause of morbidity and mortality for malnourished individuals, with infection during malnutrition representing the leading cause of childhood mortality worldwide. In the face of this complex challenge, simple refeeding protocols have remained the primary treatment strategy. Although an association between undernutrition and infection susceptibility has been appreciated for over a century, the underlying mechanisms remain poorly understood and the extent to which refeeding intervention is sufficient to reverse nutritionally acquired immunodeficiency is unclear. Here we investigate how malnutrition leads to immune dysfunction and the ability of refeeding to repair it. We find that chronic malnutrition severely impairs the ability of animals to control a sub-lethal bacterial infection. Malnourished animals exhibit blunted immune cell expansion, impaired immune function, and accelerated contraction prior to pathogen clearance. While this defect is global, we find that myelopoiesis is uniquely impacted, resulting in in reduced neutrophil and monocyte numbers prior to and post-infection. Upon refeeding, we observe that animals recover body mass, size, cellularity across all major immune organs, the capacity to undergo normal immune cell expansion in response to infection, and a restoration in T cell responses. Despite this broad improvement, refed animals remain susceptible to bacterial infection, uncoupling global lymphoid atrophy from immunodeficiency. Mechanistically, we find peripheral neutrophil and monocyte numbers fail to fully recover and refed animals are unable to undergo normal emergency myelopoiesis. Altogether, this work identifies a novel cellular link between prior nutritional state and immunocompetency, highlighting dysregulated myelopoiesis as a major driver. We believe these findings illustrate how exposure to food scarcity is an immunologic variable, even post-recovery, which should be accounted for in patient medical history and current global public health policy.
Collapse
Affiliation(s)
- Alisa Sukhina
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia; Philadelphia, PA 19104
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA 19104
| | - Clemence Queriault
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia; Philadelphia, PA 19104
| | - Elise Hall
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia; Philadelphia, PA 19104
| | - Kelly Rome
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia; Philadelphia, PA 19104
| | - Muskaan Aggarwal
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA 19104
| | - Elizabeth Nunn
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA; Department of Physiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Ashley Weiss
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia; Philadelphia, PA 19104
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA 19104
| | - Janet Nguyen
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia; Philadelphia, PA 19104
| | - Will Bailis
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia; Philadelphia, PA 19104
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA 19104
| |
Collapse
|
3
|
Do KK, Wang F, Sun X, Zhang Y, Liang W, Liu JY, Jiang DY, Lu X, Wang W, Zhang L, Dean DC, Liu Y. Conditional deletion of Zeb1 in Csf1r + cells reduces inflammatory response of the cornea to alkali burn. iScience 2024; 27:109694. [PMID: 38660397 PMCID: PMC11039400 DOI: 10.1016/j.isci.2024.109694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 01/29/2024] [Accepted: 04/05/2024] [Indexed: 04/26/2024] Open
Abstract
ZEB1 is an essential factor in embryonic development. In adults, it is often highly expressed in malignant tumors with low expression in normal tissues. The major biological function of ZEB1 in developing embryos and progressing cancers is to transdifferentiate cells from an epithelial to mesenchymal phenotype; but what roles ZEB1 plays in normal adult tissues are largely unknown. We previously reported that the reduction of Zeb1 in monoallelic global knockout (Zeb1+/-) mice reduced corneal inflammation-associated neovascularization following alkali burn. To uncover the cellular mechanism underlying the Zeb1 regulation of corneal inflammation, we functionally deleted Zeb1 alleles in Csf1r+ myeloid cells using a conditional knockout (cKO) strategy and found that Zeb1 cKO reduced leukocytes in the cornea after alkali burn. The reduction of immune cells was due to their increased apoptotic rate and linked to a Zeb1-downregulated apoptotic pathway. We conclude that Zeb1 facilitates corneal inflammatory response by maintaining Csf1r+ cell viability.
Collapse
Affiliation(s)
- Khoi K. Do
- Department of Medicine, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Fuhua Wang
- Department of Medicine, University of Louisville School of Medicine, Louisville, KY 40202, USA
- Eye Institute and Eye Hospital of Shangdong First Medical University, Jinan 250021, China
| | - Xiaolei Sun
- Department of Medicine, University of Louisville School of Medicine, Louisville, KY 40202, USA
- Eye Institute and Eye Hospital of Shangdong First Medical University, Jinan 250021, China
| | - Yingnan Zhang
- Department of Medicine, University of Louisville School of Medicine, Louisville, KY 40202, USA
- The Rosenberg School of Optometry, University of the Incarnate Word, San Antonio, TX 78229, USA
| | - Wei Liang
- Department of Medicine, University of Louisville School of Medicine, Louisville, KY 40202, USA
- Department of Ophthalmology, Third People’s Hospital of Dalian, Dalian Medical University, Dalian 116033, China
| | - John Y. Liu
- Department of Medicine, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Daniel Y. Jiang
- Department of Medicine, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Xiaoqin Lu
- Department of Medicine, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Wei Wang
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Lijun Zhang
- Department of Ophthalmology, Third People’s Hospital of Dalian, Dalian Medical University, Dalian 116033, China
| | - Douglas C. Dean
- Department of Medicine, University of Louisville School of Medicine, Louisville, KY 40202, USA
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, Louisville, KY 40202, USA
- James Brown Cancer Center, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Yongqing Liu
- Department of Medicine, University of Louisville School of Medicine, Louisville, KY 40202, USA
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, Louisville, KY 40202, USA
- James Brown Cancer Center, University of Louisville School of Medicine, Louisville, KY 40202, USA
| |
Collapse
|
4
|
Salvi J, Andreoletti P, Audinat E, Balland E, Ben Fradj S, Cherkaoui-Malki M, Heurtaux T, Liénard F, Nédélec E, Rovère C, Savary S, Véjux A, Trompier D, Benani A. Microgliosis: a double-edged sword in the control of food intake. FEBS J 2024; 291:615-631. [PMID: 35880408 DOI: 10.1111/febs.16583] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 06/30/2022] [Accepted: 07/25/2022] [Indexed: 02/16/2024]
Abstract
Maintaining energy balance is essential for survival and health. This physiological function is controlled by the brain, which adapts food intake to energy needs. Indeed, the brain constantly receives a multitude of biological signals that are derived from digested foods or that originate from the gastrointestinal tract, energy stores (liver and adipose tissues) and other metabolically active organs (muscles). These signals, which include circulating nutrients, hormones and neuronal inputs from the periphery, collectively provide information on the overall energy status of the body. In the brain, several neuronal populations can specifically detect these signals. Nutrient-sensing neurons are found in discrete brain areas and are highly enriched in the hypothalamus. In turn, specialized brain circuits coordinate homeostatic responses acting mainly on appetite, peripheral metabolism, activity and arousal. Accumulating evidence shows that hypothalamic microglial cells located at the vicinity of these circuits can influence the brain control of energy balance. However, microglial cells could have opposite effects on energy balance, that is homeostatic or detrimental, and the conditions for this shift are not totally understood yet. One hypothesis relies on the extent of microglial activation, and nutritional lipids can considerably change it.
Collapse
Affiliation(s)
- Juliette Salvi
- CSGA, Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro Dijon, Université Bourgogne Franche-Comté, Dijon, France
| | - Pierre Andreoletti
- Laboratoire Bio-PeroxIL, Université Bourgogne Franche-Comté, Dijon, France
| | - Etienne Audinat
- IGF, Université de Montpellier, CNRS, Inserm, Montpellier, France
| | - Eglantine Balland
- Department of Nutrition, Dietetics and Food, School of Clinical Sciences at Monash Health, Faculty of Medicine, Nursing and Health Sciences, Monash University, Notting Hill, Australia
| | - Selma Ben Fradj
- IPMC, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, Université Côte d'Azur, Valbonne, France
| | | | - Tony Heurtaux
- Luxembourg Center of Neuropathology (LCNP), Dudelange, Luxembourg
- Department of Life Sciences and Medicine, University of Luxembourg, Belvaux, Luxembourg
| | - Fabienne Liénard
- CSGA, Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro Dijon, Université Bourgogne Franche-Comté, Dijon, France
| | - Emmanuelle Nédélec
- CSGA, Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro Dijon, Université Bourgogne Franche-Comté, Dijon, France
| | - Carole Rovère
- IPMC, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, Université Côte d'Azur, Valbonne, France
| | - Stéphane Savary
- Laboratoire Bio-PeroxIL, Université Bourgogne Franche-Comté, Dijon, France
| | - Anne Véjux
- Laboratoire Bio-PeroxIL, Université Bourgogne Franche-Comté, Dijon, France
| | - Doriane Trompier
- Laboratoire Bio-PeroxIL, Université Bourgogne Franche-Comté, Dijon, France
| | - Alexandre Benani
- CSGA, Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro Dijon, Université Bourgogne Franche-Comté, Dijon, France
| |
Collapse
|
5
|
Kim Y, Kamada N. The role of the microbiota in myelopoiesis during homeostasis and inflammation. Int Immunol 2023; 35:267-274. [PMID: 36694400 PMCID: PMC10199171 DOI: 10.1093/intimm/dxad002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 01/23/2023] [Indexed: 01/26/2023] Open
Abstract
The microbiota engages in the development and maintenance of the host immune system. The microbiota affects not only mucosal tissues where it localizes but also the distal organs. Myeloid cells are essential for host defense as first responders of the host immune system. Their generation, called myelopoiesis, is regulated by environmental signals, including commensal microbiota. Hematopoietic stem and progenitor cells in bone marrow can directly or indirectly sense microbiota-derived signals, thereby giving rise to myeloid cell lineages at steady-state and during inflammation. In this review, we discuss the role of commensal microorganisms in the homeostatic regulation of myelopoiesis in the bone marrow. We also outline the effects of microbial signals on myelopoiesis during inflammation and infection, with a particular focus on the development of innate immune memory. Studying the relationship between the microbiota and myelopoiesis will help us understand how the microbiota regulates immune responses at a systemic level beyond the local mucosa.
Collapse
Affiliation(s)
- Yeji Kim
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Nobuhiko Kamada
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
- Laboratory of Microbiology and Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| |
Collapse
|
6
|
Chao CJ, Zhang E, Zhao Z. Engineering cells for precision drug delivery: New advances, clinical translation, and emerging strategies. Adv Drug Deliv Rev 2023; 197:114840. [PMID: 37088403 DOI: 10.1016/j.addr.2023.114840] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/04/2023] [Accepted: 04/17/2023] [Indexed: 04/25/2023]
Abstract
Cells have emerged as a promising new form of drug delivery carriers owing to their distinguished advantages such as naturally bypassing immune recognition, intrinsic capability to navigate biological barriers, and access to hard-to-reach tissues via onboarding sensing and active motility. Over the past two decades, a large body of work has focused on understanding the ability of cell carriers to breach biological barriers and to modulate drug pharmacokinetics and pharmacodynamics. These efforts have led to the engineering of various cells for tissue-specific drug delivery. Despite exciting advances, clinical translation of cell-based drug carriers demands a thorough understanding of the pressing challenges and potential strategies to overcome them. Here, we summarize recent advances and new concepts in cell-based drug carriers and their clinical translation. We also discuss key considerations and emerging strategies to engineering the next-generation cell-based delivery technologies for more precise, targeted drug delivery.
Collapse
Affiliation(s)
- Chih-Jia Chao
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Endong Zhang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Zongmin Zhao
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois Chicago, Chicago, IL 60612, USA; Translational Oncology Program, University of Illinois Cancer Center, Chicago, IL 60612.
| |
Collapse
|
7
|
Zhang F, Xu Z, Jolly KJ. Myeloid cell-mediated drug delivery: from nanomedicine to cell therapy. Adv Drug Deliv Rev 2023; 197:114827. [PMID: 37068659 DOI: 10.1016/j.addr.2023.114827] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/23/2023] [Accepted: 04/11/2023] [Indexed: 04/19/2023]
Abstract
In the presence of tissue inflammation, injury, or cancer, myeloid cells are recruited to disease regions through a multi-step process involving myelopoiesis, chemotaxis, cell migration, and diapedesis. As an emerging drug delivery approach, cell-mediated drug delivery takes advantage of the cell recruitment process to enhance the active transport of therapeutic cargo to disease regions. In the past few decades, a variety of nano-engineering methods have emerged to enhance interactions of nanoparticles with cells of interest, which can be adapted for cell-mediated drug delivery. Moreover, the drug delivery field can benefit from the recent clinical success of cell-based therapies, which created cell-engineering methods to engineer circulating leukocytes as 'living drug delivery vehicles' to target diseased tissues. In this review, we first provide an overview of myeloid cell recruitment and discuss how various factors within this process may affect cell-mediated delivery. In the second part of this review article, we summarize the status quo of nano-engineering and cell-engineering approaches and discuss how these engineering approaches can be adapted for cell-mediated delivery. Finally, we discuss future directions of this field, pointing out key challenges in the clinical translation of cell-mediated drug delivery.
Collapse
Affiliation(s)
- Fan Zhang
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, USA; Department of Chemical Engineering, College of Engineering, University of Florida, Gainesville, FL, USA; Department of Pharmacology & Therapeutics, College of Medicine, University of Florida, Gainesville, FL, USA.
| | - Zijing Xu
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Kevon J Jolly
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, USA
| |
Collapse
|
8
|
Bancaro N, Calì B, Troiani M, Elia AR, Arzola RA, Attanasio G, Lai P, Crespo M, Gurel B, Pereira R, Guo C, Mosole S, Brina D, D'Ambrosio M, Pasquini E, Spataro C, Zagato E, Rinaldi A, Pedotti M, Di Lascio S, Meani F, Montopoli M, Ferrari M, Gallina A, Varani L, Pereira Mestre R, Bolis M, Gillessen Sommer S, de Bono J, Calcinotto A, Alimonti A. Apolipoprotein E induces pathogenic senescent-like myeloid cells in prostate cancer. Cancer Cell 2023; 41:602-619.e11. [PMID: 36868226 DOI: 10.1016/j.ccell.2023.02.004] [Citation(s) in RCA: 78] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/20/2022] [Accepted: 02/06/2023] [Indexed: 03/05/2023]
Abstract
Tumor cells promote the recruitment of immunosuppressive neutrophils, a subset of myeloid cells driving immune suppression, tumor proliferation, and treatment resistance. Physiologically, neutrophils are known to have a short half-life. Here, we report the identification of a subset of neutrophils that have upregulated expression of cellular senescence markers and persist in the tumor microenvironment. Senescent-like neutrophils express the triggering receptor expressed on myeloid cells 2 (TREM2) and are more immunosuppressive and tumor-promoting than canonical immunosuppressive neutrophils. Genetic and pharmacological elimination of senescent-like neutrophils decreases tumor progression in different mouse models of prostate cancer. Mechanistically, we have found that apolipoprotein E (APOE) secreted by prostate tumor cells binds TREM2 on neutrophils, promoting their senescence. APOE and TREM2 expression increases in prostate cancers and correlates with poor prognosis. Collectively, these results reveal an alternative mechanism of tumor immune evasion and support the development of immune senolytics targeting senescent-like neutrophils for cancer therapy.
Collapse
Affiliation(s)
- Nicolò Bancaro
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, 6900 Lugano, Switzerland
| | - Bianca Calì
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, 6900 Lugano, Switzerland
| | - Martina Troiani
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, 6900 Lugano, Switzerland
| | - Angela Rita Elia
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, 6900 Lugano, Switzerland
| | - Rydell Alvarez Arzola
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, 6900 Lugano, Switzerland
| | - Giuseppe Attanasio
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, 6900 Lugano, Switzerland
| | - Ping Lai
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, 6900 Lugano, Switzerland
| | - Mateus Crespo
- The Institute of Cancer Research and the Royal Marsden NHS Foundation Trust, London, UK
| | - Bora Gurel
- The Institute of Cancer Research and the Royal Marsden NHS Foundation Trust, London, UK
| | - Rita Pereira
- The Institute of Cancer Research and the Royal Marsden NHS Foundation Trust, London, UK
| | - Christina Guo
- The Institute of Cancer Research and the Royal Marsden NHS Foundation Trust, London, UK
| | - Simone Mosole
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, 6900 Lugano, Switzerland
| | - Daniela Brina
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, 6900 Lugano, Switzerland
| | - Mariantonietta D'Ambrosio
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, 6900 Lugano, Switzerland
| | - Emiliano Pasquini
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, 6900 Lugano, Switzerland
| | - Clarissa Spataro
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, 6900 Lugano, Switzerland
| | - Elena Zagato
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, 6900 Lugano, Switzerland
| | - Andrea Rinaldi
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, 6900 Lugano, Switzerland
| | - Mattia Pedotti
- Università della Svizzera Italiana, Faculty of Biomedical Sciences, 6900 Lugano, Switzerland; Institute for Research in Biomedicine (IRB), 6500 Bellinzona, Switzerland
| | - Simona Di Lascio
- Università della Svizzera Italiana, Faculty of Biomedical Sciences, 6900 Lugano, Switzerland; Institute of Oncology of Southern Switzerland (IOSI), Ente Ospedaliero Cantonale (EOC), Bellinzona, Switzerland
| | - Francesco Meani
- Università della Svizzera Italiana, Faculty of Biomedical Sciences, 6900 Lugano, Switzerland; Institute of Oncology of Southern Switzerland (IOSI), Ente Ospedaliero Cantonale (EOC), Bellinzona, Switzerland
| | - Monica Montopoli
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy; Veneto Institute of Molecular Medicine, Padova, Italy
| | - Matteo Ferrari
- Department of Urology, Ente Ospedaliero Cantonale, Ospedale Regionale di Lugano - Civico USI - Università della Svizzera Italiana, Lugano, Switzerland
| | - Andrea Gallina
- Department of Urology, Ente Ospedaliero Cantonale, Ospedale Regionale di Lugano - Civico USI - Università della Svizzera Italiana, Lugano, Switzerland
| | - Luca Varani
- Institute for Research in Biomedicine (IRB), 6500 Bellinzona, Switzerland
| | - Ricardo Pereira Mestre
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, 6900 Lugano, Switzerland; Institute of Oncology of Southern Switzerland (IOSI), Ente Ospedaliero Cantonale (EOC), Bellinzona, Switzerland
| | - Marco Bolis
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, 6900 Lugano, Switzerland; Computational Oncology Unit, Department of Oncology, IRCCS Istituto di Ricerche Farmacologiche 'Mario Negri', Via Mario Negri 2, 20156 Milano, Italy
| | - Silke Gillessen Sommer
- Università della Svizzera Italiana, Faculty of Biomedical Sciences, 6900 Lugano, Switzerland; Institute of Oncology of Southern Switzerland (IOSI), Ente Ospedaliero Cantonale (EOC), Bellinzona, Switzerland
| | - Johann de Bono
- The Institute of Cancer Research and the Royal Marsden NHS Foundation Trust, London, UK
| | - Arianna Calcinotto
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, 6900 Lugano, Switzerland.
| | - Andrea Alimonti
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, 6900 Lugano, Switzerland; Institute of Oncology of Southern Switzerland (IOSI), Ente Ospedaliero Cantonale (EOC), Bellinzona, Switzerland; Veneto Institute of Molecular Medicine, Padova, Italy; Department of Medicine, University of Padova, Padova, Italy; Department of Health Sciences and Technology (D-HEST) ETH Zurich, 8093 Zurich, Switzerland.
| |
Collapse
|
9
|
Tommasi C, Pellegrino B, Diana A, Palafox Sancez M, Orditura M, Scartozzi M, Musolino A, Solinas C. The Innate Immune Microenvironment in Metastatic Breast Cancer. J Clin Med 2022; 11:jcm11205986. [PMID: 36294305 PMCID: PMC9604853 DOI: 10.3390/jcm11205986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/27/2022] [Accepted: 10/09/2022] [Indexed: 11/30/2022] Open
Abstract
The immune system plays a fundamental role in neoplastic disease. In the era of immunotherapy, the adaptive immune response has been in the spotlight whereas the role of innate immunity in cancer development and progression is less known. The tumor microenvironment influences the terminal differentiation of innate immune cells, which can explicate their pro-tumor or anti-tumor effect. Different cells are able to recognize and eliminate no self and tumor cells: macrophages, natural killer cells, monocytes, dendritic cells, and neutrophils are, together with the elements of the complement system, the principal players of innate immunity in cancer development and evolution. Metastatic breast cancer is a heterogeneous disease from the stromal, immune, and biological point of view and requires deepened exploration to understand different patient outcomes. In this review, we summarize the evidence about the role of innate immunity in breast cancer metastatic sites and the potential targets for optimizing the innate response as a novel treatment opportunity.
Collapse
Affiliation(s)
- Chiara Tommasi
- Medical Oncology and Breast Unit, University Hospital of Parma, 43126 Parma, Italy
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
- GOIRC (Gruppo Oncologico Italiano di Ricerca Clinica), 43126 Parma, Italy
- Correspondence:
| | - Benedetta Pellegrino
- Medical Oncology and Breast Unit, University Hospital of Parma, 43126 Parma, Italy
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
- GOIRC (Gruppo Oncologico Italiano di Ricerca Clinica), 43126 Parma, Italy
| | - Anna Diana
- Medical Oncology Unit, Ospedale del Mare, 80147 Naples, Italy
| | - Marta Palafox Sancez
- Tumor Heterogeneity, Metastasis and Resistance Laboratory, University of Basel, 4001 Basel, Switzerland
| | - Michele Orditura
- Division of Medical Oncology, Department of Precision Medicine, University of Campania Luigi Vanvitelli, 80131 Naples, Italy
| | - Mario Scartozzi
- Medical Oncology Department, University of Cagliari, 09042 Cagliari, Italy
| | - Antonino Musolino
- Medical Oncology and Breast Unit, University Hospital of Parma, 43126 Parma, Italy
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
- GOIRC (Gruppo Oncologico Italiano di Ricerca Clinica), 43126 Parma, Italy
| | - Cinzia Solinas
- Medical Oncology Department, University of Cagliari, 09042 Cagliari, Italy
| |
Collapse
|
10
|
Zemet R, Van den Veyver IB, Stankiewicz P. Parental mosaicism for apparent de novo genetic variants: Scope, detection, and counseling challenges. Prenat Diagn 2022; 42:811-821. [PMID: 35394072 PMCID: PMC9995893 DOI: 10.1002/pd.6144] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 04/04/2022] [Accepted: 04/04/2022] [Indexed: 11/07/2022]
Abstract
The disease burden of de novo mutations (DNMs) has been evidenced only recently when the common application of next-generation sequencing technologies enabled their reliable and affordable detection through family-based clinical exome or genome sequencing. Implementation of exome sequencing into prenatal diagnostics revealed that up to 63% of pathogenic or likely pathogenic variants associated with fetal structural anomalies are apparently de novo, primarily for autosomal dominant disorders. Apparent DNMs have been considered to primarily occur as germline or zygotic events, with consequently negligible recurrence risks. However, there is now evidence that a considerable proportion of them are in fact inherited from a parent mosaic for the variant. Here, we review the burden of DNMs in prenatal diagnostics and the influence of parental mosaicism on the interpretation of apparent DNMs and discuss the challenges with detecting and quantifying parental mosaicism and its effect on recurrence risk. We also describe new bioinformatic and technological tools developed to assess mosaicism and discuss how they improve the accuracy of reproductive risk counseling when parental mosaicism is detected.
Collapse
Affiliation(s)
- Roni Zemet
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Ignatia B Van den Veyver
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA.,Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, Texas, USA.,Texas Children's Hospital, Houston, Texas, USA
| | - Paweł Stankiewicz
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
11
|
Alshammary AF, Al-Sulaiman AM. The journey of SARS-CoV-2 in human hosts: a review of immune responses, immunosuppression, and their consequences. Virulence 2021; 12:1771-1794. [PMID: 34251989 PMCID: PMC8276660 DOI: 10.1080/21505594.2021.1929800] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 04/13/2021] [Accepted: 05/10/2021] [Indexed: 01/08/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a highly infectious viral disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Laboratory findings from a significant number of patients with COVID-19 indicate the occurrence of leukocytopenia, specifically lymphocytopenia. Moreover, infected patients can experience contrasting outcomes depending on lymphocytopenia status. Patients with resolved lymphocytopenia are more likely to recover, whereas critically ill patients with signs of unresolved lymphocytopenia develop severe complications, sometimes culminating in death. Why immunodepression manifests in patients with COVID-19 remains unclear. Therefore, the evaluation of clinical symptoms and laboratory findings from infected patients is critical for understanding the disease course and its consequences. In this review, we take a logical approach to unravel the reasons for immunodepression in patients with COVID-19. Following the footprints of the virus within host tissues, from entry to exit, we extrapolate the mechanisms underlying the phenomenon of immunodepression.
Collapse
Affiliation(s)
- Amal F. Alshammary
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | | |
Collapse
|
12
|
Shi SX, Shi K, Liu Q. Brain injury instructs bone marrow cellular lineage destination to reduce neuroinflammation. Sci Transl Med 2021; 13:13/589/eabc7029. [PMID: 33853930 DOI: 10.1126/scitranslmed.abc7029] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 08/06/2020] [Accepted: 01/16/2021] [Indexed: 12/22/2022]
Abstract
Acute brain injury mobilizes circulating leukocytes to transmigrate into the perivascular space and brain parenchyma. This process amplifies neural injury. Bone marrow hematopoiesis replenishes the exhausted peripheral leukocyte pools. However, it is not known whether brain injury influences the development of bone marrow lineages and how altered hematopoietic cell lineages affect neurological outcome. Here, we showed that bone marrow hematopoietic stem cells (HSCs) can be swiftly skewed toward the myeloid lineage in patients with intracerebral hemorrhage (ICH) and experimental ICH models. Lineage tracing revealed a predominantly augmented hematopoiesis of Ly6Clow monocytes infiltrating the ICH brain, where they generated alternatively activated macrophages and suppressed neuroinflammation and brain injury. The ICH brain uses β3-adrenergic innervation that involves cell division cycle 42 to promote bone marrow hematopoiesis of Ly6Clow monocytes, which could be further potentiated by the U.S. Food and Drug Administration-approved β3-adrenergic agonist mirabegron. Our results suggest that brain injury modulates HSC lineage development to curb distal brain inflammation, implicating the bone marrow as a unique niche for self-protective neuroimmune interaction that might be exploited to obtain therapeutic effects.
Collapse
Affiliation(s)
- Samuel X Shi
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China.,Interdisciplinary Neuroscience Graduate Program, Arizona State University, Tempe, AZ 85281, USA
| | - Kaibin Shi
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China.,National Clinical Research Center for Neurological Diseases of China, Jing-Jin Center for Neuroinflammation, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Qiang Liu
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China. .,Interdisciplinary Neuroscience Graduate Program, Arizona State University, Tempe, AZ 85281, USA
| |
Collapse
|
13
|
Lovászi M, Branco Haas C, Antonioli L, Pacher P, Haskó G. The role of P2Y receptors in regulating immunity and metabolism. Biochem Pharmacol 2021; 187:114419. [PMID: 33460626 DOI: 10.1016/j.bcp.2021.114419] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 01/07/2021] [Accepted: 01/11/2021] [Indexed: 02/07/2023]
Abstract
P2Y receptors are G protein-coupled receptors whose physiological agonists are the nucleotides ATP, ADP, UTP, UDP and UDP-glucose. Eight P2Y receptors have been cloned in humans: P2Y1R, P2Y2R, P2Y4R, P2Y6R, P2Y11R, P2Y12R, P2Y13R and P2Y14R. P2Y receptors are expressed in lymphoid tissues such as thymus, spleen and bone marrow where they are expressed on lymphocytes, macrophages, dendritic cells, neutrophils, eosinophils, mast cells, and platelets. P2Y receptors regulate many aspects of immune cell function, including phagocytosis and killing of pathogens, antigen presentation, chemotaxis, degranulation, cytokine production, and lymphocyte activation. Consequently, P2Y receptors shape the course of a wide range of infectious, autoimmune, and inflammatory diseases. P2Y12R ligands have already found their way into the therapeutic arena, and we envision additional ligands as future drugs for the treatment of diseases caused by or associated with immune dysregulation.
Collapse
Affiliation(s)
- Marianna Lovászi
- Department of Anesthesiology, Columbia University, New York, NY, USA
| | | | - Luca Antonioli
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Pál Pacher
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institutes of Health/NIAAA, Bethesda, MD, USA
| | - György Haskó
- Department of Anesthesiology, Columbia University, New York, NY, USA.
| |
Collapse
|
14
|
Kulikauskaite J, Wack A. Teaching Old Dogs New Tricks? The Plasticity of Lung Alveolar Macrophage Subsets. Trends Immunol 2020; 41:864-877. [PMID: 32896485 PMCID: PMC7472979 DOI: 10.1016/j.it.2020.08.008] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 08/07/2020] [Accepted: 08/07/2020] [Indexed: 12/24/2022]
Abstract
Alveolar macrophages (AMs) are highly abundant lung cells with important roles in homeostasis and immunity. Their function influences the outcome of lung infections, lung cancer, and chronic inflammatory disease. Recent findings reveal functional heterogeneity of AMs. Following lung insult, resident AMs can either remain unchanged, acquire new functionality, or be replaced by monocyte-derived AMs. Evidence from mouse models correlates AM function with their embryonic or monocyte origin. We hypothesize that resident AMs are terminally differentiated cells with low responsiveness and limited plasticity, while recruited, monocyte-derived AMs are initially highly immunoreactive but more plastic, able to change their function in response to environmental cues. Understanding cell-intrinsic and -extrinsic mechanisms determining AM function may provide opportunities for intervention in lung disease.
Collapse
Affiliation(s)
| | - Andreas Wack
- Immunoregulation Laboratory, Francis Crick Institute, London, UK.
| |
Collapse
|
15
|
Ibarra LE, Beaugé L, Arias-Ramos N, Rivarola VA, Chesta CA, López-Larrubia P, Palacios RE. Trojan horse monocyte-mediated delivery of conjugated polymer nanoparticles for improved photodynamic therapy of glioblastoma. Nanomedicine (Lond) 2020; 15:1687-1707. [DOI: 10.2217/nnm-2020-0106] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Aim: To assess monocyte-based delivery of conjugated polymer nanoparticles (CPNs) for improved photodynamic therapy (PDT) in glioblastoma (GBM). Materials & methods: Human monocyte cells (THP-1) and murine monocytes isolated from bone marrow (mBMDMs) were employed as stealth CPN carriers to penetrate into GBM spheroids and an orthotopic model of the tumor. The success of PDT, using this cell-mediated targeting strategy, was determined by its effect on the spheroids. Results: CPNs did not affect monocyte viability in the absence of light and did not show nonspecific release after cell loading. Activated monocytes incorporated CPNs in a higher proportion than monocytes in their naive state, without a loss of cellular functionality. In vitro PDT efficacy using cell-mediated delivery was superior to that using non vehiculized CPNs. Conclusion: CPN-loaded monocytes could efficiently deliver CPNs into GBM spheroids and the orthotopic model. Improved PDT in spheroids was confirmed using this delivery strategy.
Collapse
Affiliation(s)
- Luis E Ibarra
- Instituto de Biotecnología Ambiental y Salud (INBIAS), Universidad Nacional de Río Cuarto (UNRC) y Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Río Cuarto, 5800, Córdoba, Argentina
- Departamento de Biología Molecular, Facultad de Ciencias Exactas Fisicoquímicas y Naturales, UNRC, Río Cuarto, 5800, Córdoba, Argentina
| | - Lucía Beaugé
- Departamento de Biología Molecular, Facultad de Ciencias Exactas Fisicoquímicas y Naturales, UNRC, Río Cuarto, 5800, Córdoba, Argentina
| | - Nuria Arias-Ramos
- Instituto de Investigaciones Biomédicas “Alberto Sols”, CSIC/UAM, Madrid, 28029, España
| | - Viviana A Rivarola
- Instituto de Biotecnología Ambiental y Salud (INBIAS), Universidad Nacional de Río Cuarto (UNRC) y Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Río Cuarto, 5800, Córdoba, Argentina
- Departamento de Biología Molecular, Facultad de Ciencias Exactas Fisicoquímicas y Naturales, UNRC, Río Cuarto, 5800, Córdoba, Argentina
| | - Carlos A Chesta
- Instituto de Investigaciones en Tecnologías Energéticas y Materiales Avanzados (IITEMA), Universidad Nacional de Río Cuarto (UNRC) y CONICET, Río Cuarto, 5800, Córdoba, Argentina
- Departamento de Química, Facultad de Ciencias Exactas Fisicoquímicas y Naturales, UNRC, Río Cuarto, 5800, Córdoba, Argentina
| | - Pilar López-Larrubia
- Instituto de Investigaciones Biomédicas “Alberto Sols”, CSIC/UAM, Madrid, 28029, España
| | - Rodrigo E Palacios
- Instituto de Investigaciones en Tecnologías Energéticas y Materiales Avanzados (IITEMA), Universidad Nacional de Río Cuarto (UNRC) y CONICET, Río Cuarto, 5800, Córdoba, Argentina
- Departamento de Química, Facultad de Ciencias Exactas Fisicoquímicas y Naturales, UNRC, Río Cuarto, 5800, Córdoba, Argentina
| |
Collapse
|
16
|
Brostjan C, Oehler R. The role of neutrophil death in chronic inflammation and cancer. Cell Death Discov 2020; 6:26. [PMID: 32351713 PMCID: PMC7176663 DOI: 10.1038/s41420-020-0255-6] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 03/13/2020] [Accepted: 03/23/2020] [Indexed: 02/06/2023] Open
Abstract
The lifespan of a neutrophil is short and limited by programmed cell death, followed by efferocytosis. When activated or exposed to insult, neutrophil death may be delayed to support neutrophil effector functions such as phagocytosis, cytokine release, and pathogen destruction by degranulation. However, neutrophils may also alter the type of cell death and thereby affect inflammatory responses and tissue remodeling. This review briefly introduces the various forms of neutrophil death including apoptosis, necrosis/necroptosis, and the formation of so-called "neutrophil extracellular traps" (NETs), and it summarizes the clearance of dead cells by efferocytosis. Importantly, distinct types of neutrophil death have been found to drive chronic inflammatory disorders and cancer. Thus, the tumor and its microenvironment can delay neutrophil apoptosis to exploit their pro-angiogenic and pro-metastatic properties. Conversely, neutrophils may enter rapid and suicidal cell death by forming extracellular traps, which are expelled DNA strands with neutrophil proteins. Components of these DNA-protein complexes such as histones, high-mobility group protein B1, or neutrophil elastase have been found to promote cancer cell proliferation, adhesion, migration, invasion, and thereby tumor metastasis. In other settings of chronic inflammatory disease such as gout, NETs have been found protective rather than detrimental, as they promoted the local degradation of pro-inflammatory cytokines by neutrophil proteases. Thus, the interaction of neutrophils with the tissue environment extends beyond the stage of the living cell and the type of neutrophil death shapes immune responses and tissue remodeling in health and disease.
Collapse
Affiliation(s)
| | - Rudolf Oehler
- Department of Surgery, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
17
|
Figueroa-Romero C, Guo K, Murdock BJ, Paez-Colasante X, Bassis CM, Mikhail KA, Pawlowski KD, Evans MC, Taubman GF, McDermott AJ, O'Brien PD, Savelieff MG, Hur J, Feldman EL. Temporal evolution of the microbiome, immune system and epigenome with disease progression in ALS mice. Dis Model Mech 2019; 13:dmm041947. [PMID: 31597644 PMCID: PMC6906635 DOI: 10.1242/dmm.041947] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 10/05/2019] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a terminal neurodegenerative disease. Genetic predisposition, epigenetic changes, aging and accumulated life-long environmental exposures are known ALS risk factors. The complex and dynamic interplay between these pathological influences plays a role in disease onset and progression. Recently, the gut microbiome has also been implicated in ALS development. In addition, immune cell populations are differentially expanded and activated in ALS compared to healthy individuals. However, the temporal evolution of both the intestinal flora and the immune system relative to symptom onset in ALS is presently not fully understood. To better elucidate the timeline of the various potential pathological factors, we performed a longitudinal study to simultaneously assess the gut microbiome, immunophenotype and changes in ileum and brain epigenetic marks relative to motor behavior and muscle atrophy in the mutant superoxide dismutase 1 (SOD1G93A) familial ALS mouse model. We identified alterations in the gut microbial environment early in the life of SOD1G93A animals followed by motor dysfunction and muscle atrophy, and immune cell expansion and activation, particularly in the spinal cord. Global brain cytosine hydroxymethylation was also altered in SOD1G93A animals at disease end-stage compared to control mice. Correlation analysis confirmed interrelationships with the microbiome and immune system. This study serves as a starting point to more deeply comprehend the influence of gut microorganisms and the immune system on ALS onset and progression. Greater insight may help pinpoint novel biomarkers and therapeutic interventions to improve diagnosis and treatment for ALS patients.This article has an associated First Person interview with the joint first authors of the paper.
Collapse
Affiliation(s)
| | - Kai Guo
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58202, USA
| | - Benjamin J Murdock
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Christine M Bassis
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Kristen A Mikhail
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Matthew C Evans
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| | | | - Andrew J McDermott
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Phillipe D O'Brien
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Masha G Savelieff
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Junguk Hur
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58202, USA
| | - Eva L Feldman
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
18
|
Sivakumar R, Chan M, Shin JS, Nishida-Aoki N, Kenerson HL, Elemento O, Beltran H, Yeung R, Gujral TS. Organotypic tumor slice cultures provide a versatile platform for immuno-oncology and drug discovery. Oncoimmunology 2019; 8:e1670019. [PMID: 31741771 PMCID: PMC6844320 DOI: 10.1080/2162402x.2019.1670019] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Revised: 08/31/2019] [Accepted: 09/13/2019] [Indexed: 12/30/2022] Open
Abstract
Organotypic tumor slices represent a physiologically-relevant culture system for studying the tumor microenvironment. Systematic characterization of the tumor slice culture system will enable its effective application for translational research. Here, using flow cytometry-based immunophenotyping, we performed a comprehensive characterization of the immune cell composition in organotypic tumor slices prepared from four syngeneic mouse tumor models and a human liver tumor. We found that the immune cell compositions of organotypic tumor slices prepared on the same day as the tumor cores were harvested are similar. Differences were primarily observed in the lymphocyte population of a clinical hepatocellular carcinoma case. Viable populations of immune cells persisted in the tumor slices for 7 days. Despite some changes in the immune cell populations, we showed the utility of mouse tumor slices for assessing responses to immune-modulatory agents. Further, we demonstrated the ability to use patient-derived xenograft tumor slices for assessing responses to targeted and cytotoxic drugs. Overall, tumor slices provide a broadly useful platform for studying the tumor microenvironment and evaluating the preclinical efficacy of cancer therapeutics.
Collapse
Affiliation(s)
- Ramya Sivakumar
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Marina Chan
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Jiye Stella Shin
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Nao Nishida-Aoki
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Heidi L Kenerson
- Department of Surgery, University of Was hington, Seattle, WA, USA
| | - Olivier Elemento
- Englander Institute of Precision medicine, Weill Cornell Medicine, NY, USA
| | - Himisha Beltran
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Raymond Yeung
- Department of Surgery, University of Was hington, Seattle, WA, USA
| | - Taranjit S Gujral
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, USA.,Department of Pharmacology, University of Washington, Seattle, WA, USA
| |
Collapse
|
19
|
Clonal hematopoiesis of indeterminate potential and its impact on patient trajectories after stem cell transplantation. PLoS Comput Biol 2019; 15:e1006913. [PMID: 31026273 PMCID: PMC6505959 DOI: 10.1371/journal.pcbi.1006913] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 05/08/2019] [Accepted: 02/28/2019] [Indexed: 12/27/2022] Open
Abstract
Clonal hematopoiesis of indeterminate potential (CHIP) is a recently identified process where older patients accumulate distinct subclones defined by recurring somatic mutations in hematopoietic stem cells. CHIP's implications for stem cell transplantation have been harder to identify due to the high degree of mutational heterogeneity that is present within the genetically distinct subclones. In order to gain a better understanding of CHIP and the impact of clonal dynamics on transplantation outcomes, we created a mathematical model of clonal competition dynamics. Our analyses highlight the importance of understanding competition intensity between healthy and mutant clones. Importantly, we highlight the risk that CHIP poses in leading to dominance of precancerous mutant clones and the risk of donor derived leukemia. Furthermore, we estimate the degree of competition intensity and bone marrow niche decline in mice during aging by using our modeling framework. Together, our work highlights the importance of better characterizing the ecological and clonal composition in hematopoietic donor populations at the time of stem cell transplantation.
Collapse
|
20
|
Panek CA, Bruballa AC, Pineda GE, De Brasi C, Fernández-Brando RJ, Mejías MP, Ramos MV, Palermo MS. Cytokines use different intracellular mechanisms to upregulate the membrane expression of CX 3CR1 in human monocytes. Mol Immunol 2019; 108:23-33. [PMID: 30776726 DOI: 10.1016/j.molimm.2019.01.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 12/26/2018] [Accepted: 01/03/2019] [Indexed: 02/06/2023]
Abstract
Membrane expression of fractalkine (CX3CL1)-receptor (CX3CR1) is relevant in monocytes (Mo) because CX3CR1-CX3CL1 interactions might participate on both, homeostatic and pathologic conditions. We have previously demonstrated that CX3CR1 levels are decreased during culture and when Mo are differentiated into dendritic cells, but enhanced when differentiated into macrophages. Regarding soluble factors, lipopolysaccharide (LPS) accelerated the loss of CX3CR1, while interleukin (IL)-10 and Interferon-gamma (IFN-γ) prevented it. However, the comprehensive knowledge about the intracellular pathways that underlay the level of CX3CR1 expression in Mo is still incomplete. In the current work, we studied the effect of anti-inflammatory cytokines (IL-4, IL-13, IL-10), alone or together with IFN- γ on CX3CR1 expression. We found that only IL-10 and IFN-γ separately were able to prevent CX3CR1 down-modulation during culture of human Mo. Besides, Mo incubated with IL-10 plus IFN-γ showed the highest CX3CR1 expression by cell, suggesting cooperation between two different mechanism used by both cytokines. By studying intracellular mechanisms triggered by IL-10 and IFN-γ, we demonstrated that they specifically induced PI3K-dependent serine-phosphorylation of signal transducer and activator of transcription (STAT)3 or STAT1, respectively. Moreover, chemical inhibitors of STAT1 or STAT3 abrogated IFN-γ or IL-10 effects on CX3CR1 expression. Strikingly, only IL-10 increased CX3CR1 mRNA level, as consequence of augmenting mRNA stability. CX3CR1 mRNA increase was PI3K-dependent, supporting the causal link between the action of IL-10 at the CX3CR1 transcript and CX3CR1 protein level on Mo. Thus, both cytokines up-regulate CX3CR1 expression on human Mo by different intracellular mechanisms.
Collapse
Affiliation(s)
- Cecilia Analia Panek
- Laboratorio de Patogénesis e Inmunología de Procesos Infecciosos, Instituto de Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas- Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Andrea Cecilia Bruballa
- Laboratorio de Patogénesis e Inmunología de Procesos Infecciosos, Instituto de Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas- Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Gonzalo Ezequiel Pineda
- Laboratorio de Patogénesis e Inmunología de Procesos Infecciosos, Instituto de Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas- Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Carlos De Brasi
- Laboratorio de Genética Molecular de la Hemofilia, Instituto de Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas- Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Romina Jimena Fernández-Brando
- Laboratorio de Patogénesis e Inmunología de Procesos Infecciosos, Instituto de Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas- Academia Nacional de Medicina, Buenos Aires, Argentina
| | - María Pilar Mejías
- Laboratorio de Patogénesis e Inmunología de Procesos Infecciosos, Instituto de Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas- Academia Nacional de Medicina, Buenos Aires, Argentina
| | - María Victoria Ramos
- Laboratorio de Patogénesis e Inmunología de Procesos Infecciosos, Instituto de Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas- Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Marina Sandra Palermo
- Laboratorio de Patogénesis e Inmunología de Procesos Infecciosos, Instituto de Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas- Academia Nacional de Medicina, Buenos Aires, Argentina.
| |
Collapse
|
21
|
B cells and antibody production in melanoma. Mamm Genome 2018; 29:790-805. [DOI: 10.1007/s00335-018-9778-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 08/24/2018] [Indexed: 01/12/2023]
|
22
|
Taylor SJ, Duyvestyn JM, Dagger SA, Dishington EJ, Rinaldi CA, Dovey OM, Vassiliou GS, Grove CS, Langdon WY. Preventing chemotherapy-induced myelosuppression by repurposing the FLT3 inhibitor quizartinib. Sci Transl Med 2018; 9:9/402/eaam8060. [PMID: 28794285 DOI: 10.1126/scitranslmed.aam8060] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 04/19/2017] [Accepted: 06/29/2017] [Indexed: 02/06/2023]
Abstract
We describe an approach to inhibit chemotherapy-induced myelosuppression. We found that short-term exposure of mice to the FLT3 inhibitor quizartinib induced the transient quiescence of multipotent progenitors (MPPs). This property of quizartinib conferred marked protection to MPPs in mice receiving fluorouracil or gemcitabine. The protection resulted in the rapid recovery of bone marrow and blood cellularity, thus preventing otherwise lethal myelosuppression. A treatment strategy involving quizartinib priming that protected wild-type bone marrow progenitors, but not leukemic cells, from fluorouracil provided a more effective treatment than conventional induction therapy in mouse models of acute myeloid leukemia. This strategy has the potential to be extended for use in other cancers where FLT3 inhibition does not adversely affect the effectiveness of chemotherapy. Thus, the addition of quizartinib to cancer treatment regimens could markedly improve cancer patient survival and quality of life.
Collapse
Affiliation(s)
- Samuel J Taylor
- School of Pathology and Laboratory Medicine, University of Western Australia, Crawley, Western Australia 6009, Australia
| | - Johanna M Duyvestyn
- School of Pathology and Laboratory Medicine, University of Western Australia, Crawley, Western Australia 6009, Australia
| | - Samantha A Dagger
- School of Pathology and Laboratory Medicine, University of Western Australia, Crawley, Western Australia 6009, Australia
| | - Emma J Dishington
- School of Pathology and Laboratory Medicine, University of Western Australia, Crawley, Western Australia 6009, Australia
| | - Catherine A Rinaldi
- School of Pathology and Laboratory Medicine, University of Western Australia, Crawley, Western Australia 6009, Australia
| | - Oliver M Dovey
- Department of Haematology, Cambridge University Hospitals NHS Trust, Cambridge CB2 0QQ, UK
| | - George S Vassiliou
- Department of Haematology, Cambridge University Hospitals NHS Trust, Cambridge CB2 0QQ, UK.,Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - Carolyn S Grove
- School of Pathology and Laboratory Medicine, University of Western Australia, Crawley, Western Australia 6009, Australia.,PathWest Department of Haematology, Queen Elizabeth II Medical Centre, Nedlands, Western Australia 6009, Australia.,Department of Haematology, Sir Charles Gairdner Hospital, Nedlands, Western Australia 6009, Australia
| | - Wallace Y Langdon
- School of Pathology and Laboratory Medicine, University of Western Australia, Crawley, Western Australia 6009, Australia.
| |
Collapse
|
23
|
Patel VS, Ete Chan M, Rubin J, Rubin CT. Marrow Adiposity and Hematopoiesis in Aging and Obesity: Exercise as an Intervention. Curr Osteoporos Rep 2018; 16:105-115. [PMID: 29476393 PMCID: PMC5866776 DOI: 10.1007/s11914-018-0424-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW Changes in the bone marrow microenvironment, which accompany aging and obesity, including increased marrow adiposity, can compromise hematopoiesis. Here, we review deleterious shifts in molecular, cellular, and tissue activity and consider the potential of exercise to slow degenerative changes associated with aging and obesity. RECENT FINDINGS While bone marrow hematopoietic stem cells (HSC) are increased in frequency and myeloid-biased with age, the effect of obesity on HSC proliferation and differentiation remains controversial. HSC from both aged and obese environment have reduced hematopoietic reconstitution capacity following bone marrow transplant. Increased marrow adiposity affects HSC function, causing upregulation of myelopoiesis and downregulation of lymphopoiesis. Exercise, in contrast, can reduce marrow adiposity and restore hematopoiesis. The impact of marrow adiposity on hematopoiesis is determined mainly through correlations. Mechanistic studies are needed to determine a causative relationship between marrow adiposity and declines in hematopoiesis, which could aid in developing treatments for conditions that arise from disruptions in the marrow microenvironment.
Collapse
Affiliation(s)
- Vihitaben S Patel
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, 11794-2580, USA
| | - M Ete Chan
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, 11794-2580, USA
| | - Janet Rubin
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Clinton T Rubin
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, 11794-2580, USA.
| |
Collapse
|
24
|
Gibbings SL, Jakubzick CV. A Consistent Method to Identify and Isolate Mononuclear Phagocytes from Human Lung and Lymph Nodes. Methods Mol Biol 2018; 1799:381-395. [PMID: 29956166 DOI: 10.1007/978-1-4939-7896-0_28] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mononuclear phagocytes (MP) consist of macrophages, dendritic cells (DCs), and monocytes. In all organs, including the lung, there are multiple subtypes within these categories. The existence of all these cell types suggest that there is a clear division of labor and delicate balance between the MPs under steady state and inflammatory conditions. Although great strides have been made to understand MPs in the mouse lung, and human blood, little is known about the MPs that exist in the human lung and lung-draining lymph nodes (LNs), and even less is known about their functional roles, studies of which will require a large number of sorted cells. We have comprehensively examined cell surface markers previously used in a variety of organs to identify human pulmonary MPs. In the lung, we consistently identify five extravascular pulmonary MPs and three LN MPs. These MPs were present in over 100 lungs regardless of age or gender. Notably, the human blood CD141+ DCs, as described in the literature, were not observed in non-diseased lungs or their draining LNs. In the lung and draining LNs, expression of CD141 was only observed on HLADR+ CD11c+ CD14+ extravascular monocytes (often confused in the LN as resident DCs based on the level of HLADR expression and mouse LN data). In the human lung and LNs there are at least two DC subtypes expressing HLADR, DEC205 and CD1c, along with circulating monocytes that behave as either antigen-presenting cells or macrophages. Furthermore, we demonstrate how to distinguish between alveolar macrophages and interstitial macrophage subtypes. It still remains unclear how the human pulmonary MPs identified here align with mouse MPs. Clearly, we are now past the stage of cell surface marker characterization, and future studies will need to move toward understanding what these cell types are and how they function. Our hope is that the strategy described here can help the pulmonary community take this next step.
Collapse
Affiliation(s)
| | - Claudia V Jakubzick
- Department of Pediatrics, National Jewish Health, Denver, CO, USA. .,Department of Microbiology and Immunology, University of Colorado, Denver, CO, USA.
| |
Collapse
|
25
|
Gibbings SL, Jakubzick CV. Isolation and Characterization of Mononuclear Phagocytes in the Mouse Lung and Lymph Nodes. Methods Mol Biol 2018; 1809:33-44. [PMID: 29987780 DOI: 10.1007/978-1-4939-8570-8_3] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
There is a diverse population of mononuclear phagocytes (MPs) in the lungs, comprised of macrophages, dendritic cells (DCs), and monocytes. The existence of these various cell types suggests that there is a clear division of labor and delicate balance between the MPs under steady-state and inflammatory conditions. Here we describe how to identify pulmonary MPs using flow cytometry and how to isolate them via cell sorting. In steady-state conditions, murine lungs contain a uniform population of alveolar macrophages (AMs), three distinct interstitial macrophage (IM) populations, three DC subtypes, and a small number of tissue-trafficking monocytes. During an inflammatory response, the monocyte population is more abundant and complex since it acquires either macrophage-like or DC-like features. All in all, studying how these cell types interact with each other, structural cells, and other leukocytes within the environment will be important to understanding their role in maintaining homeostasis and during the development of disease.
Collapse
Affiliation(s)
| | - Claudia V Jakubzick
- Department of Pediatrics, National Jewish Health, Denver, CO, USA.
- Department of Microbiology and Immunology, University of Colorado, Denver, CO, USA.
| |
Collapse
|
26
|
Trahtemberg U, Mevorach D. Apoptotic Cells Induced Signaling for Immune Homeostasis in Macrophages and Dendritic Cells. Front Immunol 2017; 8:1356. [PMID: 29118755 PMCID: PMC5661053 DOI: 10.3389/fimmu.2017.01356] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Accepted: 10/03/2017] [Indexed: 12/24/2022] Open
Abstract
Inefficient and abnormal clearance of apoptotic cells (efferocytosis) contributes to systemic autoimmune disease in humans and mice, and inefficient chromosomal DNA degradation by DNAse II leads to systemic polyarthritis and a cytokine storm. By contrast, efficient clearance allows immune homeostasis, generally leads to a non-inflammatory state for both macrophages and dendritic cells (DCs), and contributes to maintenance of peripheral tolerance. As many as 3 × 108 cells undergo apoptosis every hour in our bodies, and one of the primary “eat me” signals expressed by apoptotic cells is phosphatidylserine (PtdSer). Apoptotic cells themselves are major contributors to the “anti-inflammatory” nature of the engulfment process, some by secreting thrombospondin-1 (TSP-1) or adenosine monophosphate and possibly other immune modulating “calm-down” signals that interact with macrophages and DCs. Apoptotic cells also produce “find me” and “tolerate me” signals to attract and immune modulate macrophages and DCs that express specific receptors for some of these signals. Neither macrophages nor DCs are uniform, and each cell type may variably express membrane proteins that function as receptors for PtdSer or for opsonins like complement or opsonins that bind to PtdSer, such as protein S and growth arrest-specific 6. Macrophages and DCs also express scavenger receptors, CD36, and integrins that function via bridging molecules such as TSP-1 or milk fat globule-EGF factor 8 protein and that differentially engage in various multi-ligand interactions between apoptotic cells and phagocytes. In this review, we describe the anti-inflammatory and pro-homeostatic nature of apoptotic cell interaction with the immune system. We do not review some forms of immunogenic cell death. We summarize the known apoptotic cell signaling events in macrophages and DCs that are related to toll-like receptors, nuclear factor kappa B, inflammasome, the lipid-activated nuclear receptors, Tyro3, Axl, and Mertk receptors, as well as induction of signal transducer and activator of transcription 1 and suppressor of cytokine signaling that lead to immune system silencing and DC tolerance. These properties of apoptotic cells are the mechanisms that enable their successful use as therapeutic modalities in mice and humans in various autoimmune diseases, organ transplantation, graft-versus-host disease, and sepsis.
Collapse
Affiliation(s)
- Uriel Trahtemberg
- General Intensive Care Unit, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Dror Mevorach
- Rheumatology Research Center, Department of Medicine, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| |
Collapse
|
27
|
Abstract
In metazoans, removal of cells in situ is involved in larval maturation, metamorphosis, and embryonic development. In adults, such cell removal plays a role in the homeostatic maintenance of cell numbers and tissue integrity as well as in the response to cell injury and damage. This removal involves uptake of the whole or fragmented target cells into phagocytes. Depending on the organism, these latter may be near-neighbor tissue cells and/or professional phagocytes such as, in vertebrates, members of the myeloid family of cells, especially macrophages. The uptake processes appear to involve specialized and highly conserved recognition ligands and receptors, intracellular signaling in the phagocytes, and mechanisms for ingestion. The recognition of cells destined for this form of removal is critical and, significantly, is distinguished for the most part from the recognition of foreign materials and organisms by the innate and adaptive immune systems. In keeping with the key role of cell removal in maintaining tissue homeostasis, constant cell removal is normally silent, i.e., does not initiate a local tissue reaction. This article discusses these complex and wide-ranging processes in general terms as well as the implications when these processes are disrupted in inflammation, immunity, and disease.
Collapse
Affiliation(s)
- Peter M Henson
- Department of Pediatrics, National Jewish Health, and Departments of Immunology and Medicine, University of Colorado, Denver, Colorado 80206;
| |
Collapse
|
28
|
Decote-Ricardo D, Nunes MP, Morrot A, Freire-de-Lima CG. Implication of Apoptosis for the Pathogenesis of Trypanosoma cruzi Infection. Front Immunol 2017; 8:518. [PMID: 28536576 PMCID: PMC5422484 DOI: 10.3389/fimmu.2017.00518] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 04/18/2017] [Indexed: 12/24/2022] Open
Abstract
Apoptosis is induced during the course of immune response to different infectious agents, and the ultimate fate is the recognition and uptake of apoptotic bodies by neighboring cells or by professional phagocytes. Apoptotic cells expose specific ligands to a set of conserved receptors expressed on macrophage cellular surface, which are the main cells involved in the clearance of the dying cells. These scavenger receptors, besides triggering the production of anti-inflammatory factors, also block the production of inflammatory mediators by phagocytes. Experimental infection of mice with the parasite Trypanosoma cruzi shows many pathological changes that parallels the evolution of human infection. Leukocytes undergoing intense apoptotic death are observed during the immune response to T. cruzi in the mouse model of the disease. T. cruzi replicate intensely and secrete molecules with immunomodulatory activities that interfere with T cell-mediated immune responses and secretion of pro-inflammatory cytokine secretion. This mechanism of immune evasion allows the infection to be established in the vertebrate host. Under inflammatory conditions, efferocytosis of apoptotic bodies generates an immune-regulatory phenotype in phagocytes, which is conducive to intracellular pathogen replication. However, the relevance of cellular apoptosis in the pathology of Chagas’ disease requires further studies. Here, we review the evidence of leukocyte apoptosis in T. cruzi infection and its immunomodulatory mechanism for disease progression.
Collapse
Affiliation(s)
- Débora Decote-Ricardo
- Instituto de Veterinária, Universidade Federal Rural do Rio de Janeiro, Seropédica, Brazil
| | - Marise P Nunes
- Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Alexandre Morrot
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Celio G Freire-de-Lima
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
29
|
Abstract
Macrophages are present in mammals from midgestation, contributing to physiologic homeostasis throughout life. Macrophages arise from yolk sac and foetal liver progenitors during embryonic development in the mouse and persist in different organs as heterogeneous, self-renewing tissue-resident populations. Bone marrow-derived blood monocytes are recruited after birth to replenish tissue-resident populations and to meet further demands during inflammation, infection and metabolic perturbations. Macrophages of mixed origin and different locations vary in replication and turnover, but are all active in mRNA and protein synthesis, fulfilling organ-specific and systemic trophic functions, in addition to host defence. In this review, we emphasise selected properties and non-immune functions of tissue macrophages which contribute to physiologic homeostasis.
Collapse
|