Network


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

Hotspot


Dive into the research topics where Alexei V. Tumanov is active.

Publication


Featured researches published by Alexei V. Tumanov.


Nature | 2010

CD95 promotes tumour growth

Lina Chen; Sun Mi Park; Alexei V. Tumanov; Annika Hau; Kenjiro Sawada; Christine Feig; Jerrold R. Turner; Yang-Xin Fu; Iris L. Romero; Ernst Lengyel; Marcus E. Peter

CD95 (also called Fas and APO-1) is a prototypical death receptor that regulates tissue homeostasis mainly in the immune system through the induction of apoptosis. During cancer progression CD95 is frequently downregulated or cells are rendered apoptosis resistant, raising the possibility that loss of CD95 is part of a mechanism for tumour evasion. However, complete loss of CD95 is rarely seen in human cancers and many cancer cells express large quantities of CD95 and are highly sensitive to CD95-mediated apoptosis in vitro. Furthermore, cancer patients frequently have elevated levels of the physiological ligand for CD95, CD95L. These data raise the possibility that CD95 could actually promote the growth of tumours through its non-apoptotic activities. Here we show that cancer cells in general, regardless of their CD95 apoptosis sensitivity, depend on constitutive activity of CD95, stimulated by cancer-produced CD95L, for optimal growth. Consistently, loss of CD95 in mouse models of ovarian cancer and liver cancer reduces cancer incidence as well as the size of the tumours. The tumorigenic activity of CD95 is mediated by a pathway involving JNK and Jun. These results demonstrate that CD95 has a growth-promoting role during tumorigenesis and indicate that efforts to inhibit its activity rather than to enhance it should be considered during cancer therapy.


Immunity | 2002

Distinct Role of Surface Lymphotoxin Expressed by B Cells in the Organization of Secondary Lymphoid Tissues

Alexei V. Tumanov; Dmitry V. Kuprash; Maria A. Lagarkova; Sergei I. Grivennikov; Koichiro Abe; Alexander N. Shakhov; Ludmila N. Drutskaya; Colin L. Stewart; Alexander V. Chervonsky; Sergei A. Nedospasov

In order to definitively ascertain the functional contribution of lymphotoxin (LT) expressed by B cells, we produced mice with the LTbeta gene deleted from B cells (B-LTbeta KO mice). In contrast to systemic LTbeta deletion, in B-LTbeta KO mice only splenic microarchitecture was affected, while lymph nodes and Peyers patches (PP) were normal, except for PPs reduced size. Even though B-LTbeta KO spleens retained a small number of follicular dendritic cells (FDC) which appeared to be dependent on LTbeta produced by T cells, IgG responses to sheep red blood cells were markedly reduced. Thus, the organogenic function of B-LTbeta is almost entirely restricted to spleen, where it supports the correct lymphoid architecture that is critical for an effective humoral immune response.


Science | 2013

Nonredundant Function of Soluble LTα3 Produced by Innate Lymphoid Cells in Intestinal Homeostasis

Andrey A. Kruglov; Sergei I. Grivennikov; Dmitry V. Kuprash; Caroline Winsauer; Sandra Prepens; Gitta Maria Seleznik; Gerard Eberl; Dan R. Littman; Mathias Heikenwalder; Alexei V. Tumanov; Sergei A. Nedospasov

Command and Control Innate lymphoid cells are vital for the development of gut-associated lymphoid tissues, maintenance of the epithelial barrier, and protection against intestinal microbes; their dysfunction can promote immune pathology. Immunoglobulin A (IgA) production is important for maintenance of the gut epithelial barrier and the composition of the gut microbiota. Through the generation of knockout mouse models, Kruglov et al. (p. 1243) were able to distinguish how soluble and membrane-bound lymphotoxins expressed by innate lymphoid cells in the gut specifically regulate IgA production and thereby control gut microbiota composition. Soluble lymphotoxin plays a paracrine role in controlling immunoglobulin A responses and regulating gut microbiota. Immunoglobulin A (IgA) production at mucosal surfaces contributes to protection against pathogens and controls intestinal microbiota composition. However, mechanisms regulating IgA induction are not completely defined. We show that soluble lymphotoxin α (sLTα3) produced by RORγt+ innate lymphoid cells (ILCs) controls T cell–dependent IgA induction in the lamina propria via regulation of T cell homing to the gut. By contrast, membrane-bound lymphotoxin β (LTα1β2) produced by RORγt+ ILCs is critical for T cell–independent IgA induction in the lamina propria via control of dendritic cell functions. Ablation of LTα in RORγt+ cells abrogated IgA production in the gut and altered microbiota composition. Thus, soluble and membrane-bound lymphotoxins produced by ILCs distinctly organize adaptive immune responses in the gut and control commensal microbiota composition.


Immunity | 2010

Lymphotoxin Beta Receptor Signaling in Intestinal Epithelial Cells Orchestrates Innate Immune Responses against Mucosal Bacterial Infection

Yugang Wang; Ekaterina P. Koroleva; Andrei A. Kruglov; Dmitry V. Kuprash; Sergei A. Nedospasov; Yang-Xin Fu; Alexei V. Tumanov

Epithelial cells provide the first line of defense against mucosal pathogens; however, their coordination with innate and adaptive immune cells is not well understood. Using mice with conditional gene deficiencies, we found that lymphotoxin (LT) from innate cells expressing transcription factor RORgammat, but not from adaptive T and B cells, was essential for the control of mucosal C. rodentium infection. We demonstrate that the LTbetaR signaling was required for the regulation of the early innate response against infection. Furthermore, we have revealed that LTbetaR signals in gut epithelial cells and hematopoietic-derived cells coordinate to protect the host from infection. We further determined that LTbetaR signaling in intestinal epithelial cells was required for recruitment of neutrophils to the infection site early during infection via production of CXCL1 and CXCL2 chemokines. These results support a model wherein LT from RORgammat(+) cells orchestrates the innate immune response against mucosal microbial infection.


Immunological Reviews | 2003

Dissecting the role of lymphotoxin in lymphoid organs by conditional targeting

Alexei V. Tumanov; Sergei I. Grivennikov; Alexander N. Shakhov; Stanislav A. Rybtsov; Ekaterina P. Koroleva; Junji Takeda; Sergei A. Nedospasov; Dmitry V. Kuprash

Summary:  Mice with inactivation of lymphotoxin β receptor (LTβR) system have profound defects in the development and maintenance of peripheral lymphoid organs. As surface LT is expressed by lymphocytes, natural killer cells, and lymphoid tissue‐initiating cells as well as by some other cell types, we dissected cell type‐specific LT contribution into the complex LT‐deficient phenotype by conditional gene targeting. B‐LTβ knockout (KO) mice displayed an intermediate phenotype in spleen as compared with mice with complete LTβ deficiency. In contrast, T‐LTβ KO mice displayed normal structure of the spleen. However, inactivation of LTβ in both T and B cells resulted in additional defects in the structure of the marginal zone and in the development of follicular dendritic cells in spleen. Structure of lymph nodes (LN) and Peyers patches (PP) was normal in both B‐LTβ KO and T‐ and B‐LTβ KO mice, except that PPs were of reduced size. When compared across the panel of lymphocyte‐specific LT KOs, the defects in antibody responses to T‐cell‐dependent antigens correlated with the severity of defects in spleen structure. Expression of CCL21 and CCL19 chemokines was not affected in spleen, LN and PP of B‐LTβ KO and T‐ and B‐LTβ KO mice, while CXCL13 was slightly reduced only in spleen. Collectively, our data suggest the following: (i) requirements for LT signaling to support architecture of spleen, LN and PP are different; (ii) LT complex expressed by B cells plays a major role in the maintenance of spleen structure, while surface LT expressed by T cells provides a complementary but distinct signal; and (iii) in a non‐transgenic model, expression of lymphoid tissue chemokines is only minimally dependent on the expression of surface LT complex on B and T lymphocytes.


Immunity | 2012

B Cell Maintenance of Subcapsular Sinus Macrophages Protects against a Fatal Viral Infection Independent of Adaptive Immunity

E. Ashley Moseman; Matteo Iannacone; Lidia Bosurgi; Elena Tonti; Nicolas Chevrier; Alexei V. Tumanov; Yang-Xin Fu; Nir Hacohen; Ulrich H. von Andrian

Neutralizing antibodies have been thought to be required for protection against acutely cytopathic viruses, such as the neurotropic vesicular stomatitis virus (VSV). Utilizing mice that possess B cells but lack antibodies, we show here that survival upon subcutaneous (s.c.) VSV challenge was independent of neutralizing antibody production or cell-mediated adaptive immunity. However, B cells were absolutely required to provide lymphotoxin (LT) α1β2, which maintained a protective subcapsular sinus (SCS) macrophage phenotype within virus draining lymph nodes (LNs). Macrophages within the SCS of B cell-deficient LNs, or of mice that lack LTα1β2 selectively in B cells, displayed an aberrant phenotype, failed to replicate VSV, and therefore did not produce type I interferons, which were required to prevent fatal VSV invasion of intranodal nerves. Thus, although B cells are essential for survival during VSV infection, their contribution involves the provision of innate differentiation and maintenance signals to macrophages, rather than adaptive immune mechanisms.


Nature Immunology | 2012

Lymphotoxin regulates commensal responses to enable diet-induced obesity

Vaibhav Upadhyay; Valeriy Poroyko; Tae Jin Kim; Suzanne Devkota; Sherry Fu; Donald C. Liu; Alexei V. Tumanov; Ekaterina P. Koroleva; Liufu Deng; Cathryn R. Nagler; Eugene B. Chang; Hong Tang; Yang-Xin Fu

Microbiota are essential for weight gain in mouse models of diet-induced obesity (DIO), but the pathways that cause the microbiota to induce weight gain are unknown. We report that mice deficient in lymphotoxin, a key molecule in gut immunity, were resistant to DIO. Ltbr−/− mice had different microbial community composition compared to their heterozygous littermates, including an overgrowth of segmented filamentous bacteria (SFB). Furthermore, cecal transplantation conferred leanness to germ-free recipients. Housing Ltbr−/− mice with their obese siblings rescued weight gain in Ltbr−/− mice, demonstrating the communicability of the obese phenotype. Ltbr−/− mice lacked interleukin 23 (IL-23) and IL-22, which can regulate SFB. Mice deficient in these pathways also resisted DIO, demonstrating that intact mucosal immunity guides diet-induced changes to the microbiota to enable obesity.


Cytokine & Growth Factor Reviews | 2008

Physiological functions of tumor necrosis factor and the consequences of its pathologic overexpression or blockade: mouse models.

Andrei A. Kruglov; Anna Kuchmiy; Sergei I. Grivennikov; Alexei V. Tumanov; Dmitry V. Kuprash; Sergei A. Nedospasov

TNF is an exciting cytokine which has helped to establish many paradigms in immunology. Although TNF itself has found only very limited use in the clinic, anti-cytokine therapy, which targets this single molecule, has enjoyed astounding success in treatment of a growing number of human diseases. However, since TNF mediates unique physiologic functions, in particular those related to host defense, TNF blockade may result in unwanted consequences. Much of our understanding about TNF intrinsic functions in the body, as well as about consequences of its overexpression and ablation, is based on studying phenotypes of various genetically engineered mice. Here we review mouse studies aimed at understanding TNF physiologic functions using transgenic and knockout models, and we discuss additional mouse models that may be helpful in the future.


European Journal of Immunology | 2005

Novel tumor necrosis factor‐knockout mice that lack Peyer's patches

Dmitry V. Kuprash; Alexei V. Tumanov; Dmitry J. Liepinsh; Ekaterina P. Koroleva; Marina S. Drutskaya; Andrei A. Kruglov; Alexander N. Shakhov; Eileen Southon; William J. Murphy; Lino Tessarollo; Sergei I. Grivennikov; Sergei A. Nedospasov

We generated a novel tumor necrosis factor (TNF) null mutation using Cre‐loxP technology. Mice homozygous for this mutation differ from their “conventional” counterparts; in particular, they completely lack Peyers patches (PP) but retain all lymph nodes. Our analysis of these novel TNF‐knockout mice supports the previously disputed notion of the involvement of TNF‐TNFR1 signaling in PP organogenesis. Availability of TNF‐knockout strains both with and without PP enables more definitive studies concerning the roles of TNF and PP in various immune functions and disease conditions. Here, we report that systemic ablation of TNF, but not the presence of PP per se, is critical for protection against intestinal Listeria infection in mice.


Molecular and Cellular Biology | 2002

Redundancy in Tumor Necrosis Factor (TNF) and Lymphotoxin (LT) Signaling In Vivo: Mice with Inactivation of the Entire TNF/LT Locus versus Single-Knockout Mice

Dmitry V. Kuprash; Marat B. Alimzhanov; Alexei V. Tumanov; Sergei I. Grivennikov; Alexander N. Shakhov; Ludmila N. Drutskaya; Michael W. Marino; Regina L. Turetskaya; Arthur O. Anderson; Klaus Rajewsky; Klaus Pfeffer; Sergei A. Nedospasov

ABSTRACT Homologous genes and gene products often have redundant physiological functions. Members of the tumor necrosis factor (TNF) family of cytokines can signal activation, proliferation, differentiation, costimulation, inhibition, or cell death, depending on the type and status of the target cell. TNF, lymphotoxin α (LTα), and LTβ form a subfamily of a larger family of TNF-related ligands with their genes being linked within a compact 12-kb cluster inside the major histocompatibility complex locus. Singly TNF-, LTα-, and LTβ-deficient mice share several phenotypic features, suggesting that TNF/LT signaling pathways may regulate overlapping sets of target genes. In order to directly address the issue of redundancy of TNF/LT signaling, we used the Cre-loxP recombination system to create mice with a deletion of the entire TNF/LT locus. Mice with a triple LTβ/TNF/LTα deficiency essentially manifest a combination of LT and TNF single-knockout phenotypes, except for microarchitecture of the spleen, where the disorder of lymphoid cell positioning and functional T- and B-cell compartmentalization is severer than that found in TNF or LT single-knockout mice. Thus, our data support the notion that TNF and LT have largely nonredundant functions in vivo.

Collaboration


Dive into the Alexei V. Tumanov's collaboration.

Top Co-Authors

Avatar

Sergei A. Nedospasov

Engelhardt Institute of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar

Dmitry V. Kuprash

Engelhardt Institute of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar

Ekaterina P. Koroleva

Engelhardt Institute of Molecular Biology

View shared research outputs
Top Co-Authors

Avatar

Yang-Xin Fu

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alexander N. Shakhov

Ludwig Institute for Cancer Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Annika Hau

Northwestern University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge