Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Dmitry M. Shayakhmetov is active.

Publication


Featured researches published by Dmitry M. Shayakhmetov.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Caspase-8 and RIP kinases regulate bacteria-induced innate immune responses and cell death

Dan Weng; Robyn Marty-Roix; Sandhya Ganesan; Megan K. Proulx; Gregory I. Vladimer; William J. Kaiser; Edward S. Mocarski; Kimberly Pouliot; Francis Ka-Ming Chan; Michelle A. Kelliher; Phillip A. Harris; John Bertin; Peter J. Gough; Dmitry M. Shayakhmetov; Jon D. Goguen; Katherine A. Fitzgerald; Neal S. Silverman; Egil Lien

Significance Receptor-interacting protein-1 (RIP1) kinase and caspase-8 are important players in activation of apoptotic pathways. Here we show that RIP1, caspase-8, and RIP3 contribute to infection-induced macrophage cell death and also are required for activation of transcription factor NF-κB and caspase-1 upon infection with the bacterial pathogen Yersinia pestis, the causative agent of plague. Mice lacking caspase-8 and RIP3 are also very susceptible to bacterial infection. This suggests that RIP1, caspase-8, and RIP3 are key molecules with multiple roles in innate immunity during bacterial challenge. A number of pathogens cause host cell death upon infection, and Yersinia pestis, infamous for its role in large pandemics such as the “Black Death” in medieval Europe, induces considerable cytotoxicity. The rapid killing of macrophages induced by Y. pestis, dependent upon type III secretion system effector Yersinia outer protein J (YopJ), is minimally affected by the absence of caspase-1, caspase-11, Fas ligand, and TNF. Caspase-8 is known to mediate apoptotic death in response to infection with several viruses and to regulate programmed necrosis (necroptosis), but its role in bacterially induced cell death is poorly understood. Here we provide genetic evidence for a receptor-interacting protein (RIP) kinase–caspase-8-dependent macrophage apoptotic death pathway after infection with Y. pestis, influenced by Toll-like receptor 4-TIR-domain-containing adapter-inducing interferon-β (TLR4-TRIF). Interestingly, macrophages lacking either RIP1, or caspase-8 and RIP3, also had reduced infection-induced production of IL-1β, IL-18, TNF, and IL-6; impaired activation of the transcription factor NF-κB; and greatly compromised caspase-1 processing. Cleavage of the proform of caspase-1 is associated with triggering inflammasome activity, which leads to the maturation of IL-1β and IL-18, cytokines important to host responses against Y. pestis and many other infectious agents. Our results identify a RIP1–caspase-8/RIP3-dependent caspase-1 activation pathway after Y. pestis challenge. Mice defective in caspase-8 and RIP3 were also highly susceptible to infection and displayed reduced proinflammatory cytokines and myeloid cell death. We propose that caspase-8 and the RIP kinases are key regulators of macrophage cell death, NF-κB and inflammasome activation, and host resistance after Y. pestis infection.


Nature Immunology | 2016

Interleukin 1[alpha] and the inflammatory process

Nelson C. Di Paolo; Dmitry M. Shayakhmetov

Inflammation occurs after disruption of tissue homeostasis by cell stress, injury or infection and ultimately involves the recruitment and retention of cells of hematopoietic origin, which arrive at the affected sites to resolve damage and initiate repair. Interleukin 1α (IL-1α) and IL-1β are equally potent inflammatory cytokines that activate the inflammatory process, and their deregulated signaling causes devastating diseases manifested by severe acute or chronic inflammation. Although much attention has been given to understanding the biogenesis of IL-1β, the biogenesis of IL-1α and its distinctive role in the inflammatory process remain poorly defined. In this review we examine key aspects of IL-1α biology and regulation and discuss its emerging importance in the initiation and maintenance of inflammation that underlie the pathology of many human diseases.


Infection and Immunity | 2014

Mycobacterium tuberculosis Strains Lacking Surface Lipid Phthiocerol Dimycocerosate Are Susceptible to Killing by an Early Innate Host Response

Tracey Day; John E. Mittler; Molly R. Nixon; Cullen Thompson; Maurine D. Miner; Mark J. Hickey; Reiling P. Liao; Jennifer Pang; Dmitry M. Shayakhmetov; David R. Sherman

ABSTRACT The innate immune response plays an important but unknown role in host defense against Mycobacterium tuberculosis. To define the function of innate immunity during tuberculosis, we evaluated M. tuberculosis replication dynamics during murine infection. Our data show that the early pulmonary innate immune response limits M. tuberculosis replication in a MyD88-dependent manner. Strikingly, we found that little M. tuberculosis cell death occurs during the first 2 weeks of infection. In contrast, M. tuberculosis cells deficient in the surface lipid phthiocerol dimycocerosate (PDIM) exhibited significant death rates, and consequently, total bacterial numbers were reduced. Host restriction of PDIM-deficient M. tuberculosis was not alleviated by the absence of interferon gamma (IFN-γ), inducible nitric oxide synthase (iNOS), or the phagocyte oxidase subunit p47. Taken together, these data indicate that PDIM protects M. tuberculosis from an early innate host response that is independent of IFN-γ, reactive nitrogen intermediates, and reactive oxygen species. By employing a pathogen replication tracking tool to evaluate M. tuberculosis replication and death during infection, we identify both host and pathogen factors affecting the outcome of infection.


Immunity | 2015

Interdependence between Interleukin-1 and Tumor Necrosis Factor Regulates TNF-Dependent Control of Mycobacterium tuberculosis Infection

Nelson C. Di Paolo; Shahin Shafiani; Tracey Day; Thalia Papayannopoulou; David W. Russell; Yoichiro Iwakura; David R. Sherman; Kevin B. Urdahl; Dmitry M. Shayakhmetov

The interleukin-1 receptor I (IL-1RI) is critical for host resistance to Mycobacterium tuberculosis (Mtb), yet the mechanisms of IL-1RI-mediated pathogen control remain unclear. Here, we show that without IL-1RI, Mtb-infected newly recruited Ly6G(hi) myeloid cells failed to upregulate tumor necrosis factor receptor I (TNF-RI) and to produce reactive oxygen species, resulting in compromised pathogen control. Furthermore, simultaneous ablation of IL-1RI and TNF-RI signaling on either stroma or hematopoietic cells led to early lethality, indicating non-redundant and synergistic roles of IL-1 and TNF in mediating macrophage-stroma cross-talk that was critical for optimal control of Mtb infection. Finally, we show that even in the presence of functional Mtb-specific adaptive immunity, the lack of IL-1α and not IL-1β led to an exuberant intracellular pathogen replication and progressive non-resolving inflammation. Our study reveals functional interdependence between IL-1 and TNF in enabling Mtb control mechanisms that are critical for host survival.


PLOS Pathogens | 2014

IL-1α and Complement Cooperate in Triggering Local Neutrophilic Inflammation in Response to Adenovirus and Eliminating Virus-Containing Cells

Nelson C. Di Paolo; Lisa K. Baldwin; Eric E. Irons; Thalia Papayannopoulou; Stephen Tomlinson; Dmitry M. Shayakhmetov

Inflammation is a highly coordinated host response to infection, injury, or cell stress. In most instances, the inflammatory response is pro-survival and is aimed at restoring physiological tissue homeostasis and eliminating invading pathogens, although exuberant inflammation can lead to tissue damage and death. Intravascular injection of adenovirus (Ad) results in virus accumulation in resident tissue macrophages that trigger activation of CXCL1 and CXCL2 chemokines via the IL-1α-IL-1RI signaling pathway. However, the mechanistic role and functional significance of this pathway in orchestrating cellular inflammatory responses to the virus in vivo remain unclear. Resident metallophilic macrophages expressing macrophage receptor with collagenous structure (MARCO+) in the splenic marginal zone (MZ) play the principal role in trapping Ad from the blood. Here we show that intravascular Ad administration leads to the rapid recruitment of Ly-6G+7/4+ polymorphonuclear leukocytes (PMNs) in the splenic MZ, the anatomical compartment that remains free of PMNs when these cells are purged from the bone marrow via a non-inflammatory stimulus. Furthermore, PMN recruitment in the splenic MZ resulted in elimination of virus-containing cells. IL-1α-IL-1RI signaling is only partially responsible for PMN recruitment in the MZ and requires CXCR2, but not CXCR1 signaling. We further found reduced recruitment of PMNs in the splenic MZ in complement C3-deficient mice, and that pre-treatment of IL-1α-deficient, but not wild-type mice, with complement inhibitor CR2-Crry (inhibits all complement pathways at C3 activation) or CR2-fH (inhibits only the alternative complement activation pathway) prior to Ad infection, abrogates PMN recruitment to the MZ and prevents elimination of MARCO+ macrophages from the spleen. Collectively, our study reveals a non-redundant role of the molecular factors of innate immunity – the chemokine-activating IL-1α-IL-1RI-CXCR2 axis and complement – in orchestrating local inflammation and functional cooperation of PMNs and resident macrophages in the splenic MZ, which collectively contribute to limiting disseminated pathogen spread via elimination of virus-containing cells.


Gene Therapy | 2014

Retinal transduction profiles by high-capacity viral vectors

Agostina Puppo; Giulia Cesi; Elena Marrocco; Pasquale Piccolo; Sarah Jacca; Dmitry M. Shayakhmetov; Robin J. Parks; Beverly L. Davidson; Stefano Colloca; Nicola Brunetti-Pierri; Philip Ng; Gaetano Donofrio; Alberto Auricchio

Retinal gene therapy with adeno-associated viral (AAV) vectors is safe and effective in humans. However, the limited cargo capacity of AAV prevents their use for therapy of those inherited retinopathies (IRs) due to mutations in large (>5 kb) genes. Viral vectors derived from adenovirus (Ad), lentivirus (LV) and herpes virus (HV) can package large DNA sequences, but do not target efficiently retinal photoreceptors (PRs) where the majority of genes responsible for IRs are expressed. Here, we have evaluated the mouse retinal transduction profiles of vectors derived from 16 different Ad serotypes, 7 LV pseudotypes and from a bovine HV. Most of the vectors tested transduced efficiently the retinal pigment epithelium. We found that LV-GP64 tends to transduce more PRs than the canonical LV-VSVG, albeit this was restricted to a narrow region. We observed more extensive PR transduction with HdAd1, 2 and 5/F35++ than with LV, although none of them outperformed the canonical HdAd5 or matched the extension of PR transduction achieved with AAV2/8.


Journal of Virology | 2016

Hepatocyte Heparan Sulfate Is Required for Adeno-Associated Virus 2 but Dispensable for Adenovirus 5 Liver Transduction In Vivo

Anne K. Zaiss; Erin M. Foley; Roger Lawrence; Lina S. Schneider; Hamidreza Hoveida; Patrick Secrest; Arthur Catapang; Yu Yamaguchi; Ramon Alemany; Dmitry M. Shayakhmetov; Jeffrey D. Esko; Harvey R. Herschman

ABSTRACT Adeno-associated virus 2 (AAV2) and adenovirus 5 (Ad5) are promising gene therapy vectors. Both display liver tropism and are currently thought to enter hepatocytes in vivo through cell surface heparan sulfate proteoglycans (HSPGs). To test directly this hypothesis, we created mice that lack Ext1, an enzyme required for heparan sulfate biosynthesis, in hepatocytes. Ext1 HEP mutant mice exhibit an 8-fold reduction of heparan sulfate in primary hepatocytes and a 5-fold reduction of heparan sulfate in whole liver tissue. Conditional hepatocyte Ext1 gene deletion greatly reduced AAV2 liver transduction following intravenous injection. Ad5 transduction requires blood coagulation factor X (FX); FX binds to the Ad5 capsid hexon protein and bridges the virus to HSPGs on the cell surface. Ad5.FX transduction was abrogated in primary hepatocytes from Ext1 HEP mice. However, in contrast to the case with AAV2, Ad5 transduction was not significantly reduced in the livers of Ext1 HEP mice. FX remained essential for Ad5 transduction in vivo in Ext1 HEP mice. We conclude that while AAV2 requires HSPGs for entry into mouse hepatocytes, HSPGs are dispensable for Ad5 hepatocyte transduction in vivo. This study reopens the question of how adenovirus enters cells in vivo. IMPORTANCE Our understanding of how viruses enter cells, and how they can be used as therapeutic vectors to manage disease, begins with identification of the cell surface receptors to which viruses bind and which mediate viral entry. Both adeno-associated virus 2 and adenovirus 5 are currently thought to enter hepatocytes in vivo through heparan sulfate proteoglycans (HSPGs). However, direct evidence for these conclusions is lacking. Experiments presented herein, in which hepatic heparan sulfate synthesis was genetically abolished, demonstrated that HSPGs are not likely to function as hepatocyte Ad5 receptors in vivo. The data also demonstrate that HSPGs are required for hepatocyte transduction by AAV2. These results reopen the question of the identity of the Ad5 receptor in vivo and emphasize the necessity of demonstrating the nature of the receptor by genetic means, both for understanding Ad5 entry into cells in vivo and for optimization of Ad5 vectors as therapeutic agents.


PLOS ONE | 2016

Immune complexes indirectly suppress the generation of Th17 Responses In Vivo

Ceren Ciraci; John R. Janczy; Nidhi Jain; Stefanie Haasken; Cyntia Pecli e Silva; Claudia F. Benjamim; Jeffrey J. Sadler; Alicia K. Olivier; Yoichiro Iwakura; Dmitry M. Shayakhmetov; Fayyaz S. Sutterwala; Suzanne L. Cassel

The precise context in which the innate immune system is activated plays a pivotal role in the subsequent instruction of CD4+ T helper (Th) cell responses. Th1 responses are downregulated when antigen is encountered in the presence of antigen-IgG immune complexes. To assess if Th17 responses to antigen are subject to similar influences in the presence of immune complexes we utilized an inflammatory airway disease model in which immunization of mice with Complete Freund’s Adjuvant (CFA) and ovalbumin (Ova) induces a powerful Ova-specific Th1 and Th17 response. Here we show that modification of that immunization with CFA to include IgG-Ova immune complexes results in the suppression of CFA-induced Th17 responses and a concurrent enhancement of Ova-specific Th2 responses. Furthermore, we show the mechanism by which these immune complexes suppress Th17 responses is through the enhancement of IL-10 production. In addition, the generation of Th17 responses following immunization with CFA and Ova were dependent on IL-1α but independent of NLRP3 inflammasome activation. Together these data represent a novel mechanism by which the generation of Th17 responses is regulated.


Current Opinion in Virology | 2016

Adenovirus sensing by the immune system

Svetlana Atasheva; Dmitry M. Shayakhmetov

The host immune system developed multiple ways for recognition of viral pathogens. Upon disseminated adenovirus infection, the immune system senses adenovirus invasion from the moment it enters the bloodstream. The soluble blood factors, FX, antibodies, and complement, can bind and activate plethora of host-protective immune responses. Adenovirus binding to the cellular β3 integrin and endosomal membrane rupture trigger activation of IL-1α/IL-1R1 proinflammatory cascade leading to attraction of cytotoxic immune cells to the site of infection. Upon cell entry, adenovirus exposes its DNA genome in the cytoplasm and triggers DNA sensors signaling. Even when inside the nucleus, the specialized cellular machinery that recognizes the double-strand DNA breaks become activated and triggers viral DNA replication arrest. Thus, the host employs very diverse mechanisms to prevent viral dissemination.


Immunity | 2018

Caspase-8 Collaborates with Caspase-11 to Drive Tissue Damage and Execution of Endotoxic Shock

Pratyusha Mandal; Yanjun Feng; John D. Lyons; Scott B. Berger; Shunsuke Otani; Alexandra DeLaney; Gregory K. Tharp; Kristal Maner-Smith; Eileen M. Burd; Michelle Schaeffer; Sandra J. Hoffman; Carol Capriotti; Linda Roback; Cedrick B. Young; Zhe Liang; Eric A. Ortlund; Nelson C. DiPaolo; Steven E. Bosinger; John Bertin; Peter J. Gough; Igor E. Brodsky; Craig M. Coopersmith; Dmitry M. Shayakhmetov; Edward S. Mocarski

Summary The execution of shock following high dose E. coli lipopolysaccharide (LPS) or bacterial sepsis in mice required pro‐apoptotic caspase‐8 in addition to pro‐pyroptotic caspase‐11 and gasdermin D. Hematopoietic cells produced MyD88‐ and TRIF‐dependent inflammatory cytokines sufficient to initiate shock without any contribution from caspase‐8 or caspase‐11. Both proteases had to be present to support tumor necrosis factor‐ and interferon‐&bgr;‐dependent tissue injury first observed in the small intestine and later in spleen and thymus. Caspase‐11 enhanced the activation of caspase‐8 and extrinsic cell death machinery within the lower small intestine. Neither caspase‐8 nor caspase‐11 was individually sufficient for shock. Both caspases collaborated to amplify inflammatory signals associated with tissue damage. Therefore, combined pyroptotic and apoptotic signaling mediated endotoxemia independently of RIPK1 kinase activity and RIPK3 function. These observations bring to light the relevance of tissue compartmentalization to disease processes in vivo where cytokines act in parallel to execute diverse cell death pathways. Graphical Abstract Figure. No caption available. HighlightsPro‐apoptotic Casp8 is essential for lethal LPS shock and E. coli sepsisInitiation of cytokine production proceeds independent of Casp8 and Casp11TNF and type 1 IFN drive Casp8 and Casp11 collaboration in target tissuesCombined pro‐apoptotic and pro‐pyroptotic signaling executes LPS shock &NA; Endotoxic shock requires inflammatory cytokines and cell death; however, initiation and execution signaling pathways remain unresolved. Mandal et al. describe a collaboration between pro‐apoptotic caspase‐8 and pro‐pyroptotic caspase‐11, independent of pro‐necroptotic RIPK1 kinase activity or RIPK3, to execute TNF‐ and type I interferon‐mediated inflammatory tissue damage underlying endotoxic shock.

Collaboration


Dive into the Dmitry M. Shayakhmetov's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yoichiro Iwakura

Tokyo University of Science

View shared research outputs
Top Co-Authors

Avatar

André Lieber

University of Washington

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Eric E. Irons

Roswell Park Cancer Institute

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge