Network


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

Hotspot


Dive into the research topics where Ron N. Apte is active.

Publication


Featured researches published by Ron N. Apte.


Proceedings of the National Academy of Sciences of the United States of America | 2003

IL-1 is required for tumor invasiveness and angiogenesis

Elena Voronov; Dror S. Shouval; Yakov Krelin; Emanuela Cagnano; Daniel Benharroch; Yoichiro Iwakura; Charles A. Dinarello; Ron N. Apte

Here, we describe that microenvironmental IL-1β and, to a lesser extent, IL-1α are required for in vivo angiogenesis and invasiveness of different tumor cells. In IL-1β knockout (KO) mice, local tumor or lung metastases of B16 melanoma cells were not observed compared with WT mice. Angiogenesis was assessed by the recruitment of blood vessel networks into Matrigel plugs containing B16 melanoma cells; vascularization of the plugs was present in WT mice, but was absent in IL-1β KO mice. The addition of exogenous IL-1 into B16-containing Matrigel plugs in IL-1β KO mice partially restored the angiogenic response. Moreover, the incorporation of IL-1 receptor antagonist to B16-containing plugs in WT mice inhibited the ingrowth of blood vessel networks into Matrigel plugs. In IL-1α KO mice, local tumor development and induction of an angiogenic response in Matrigel plugs was less pronounced than in WT mice, but significantly higher than in IL-1β KO mice. These effects of host-derived IL-1α and IL-1β were not restricted to the melanoma model, but were also observed in DA/3 mammary and prostate cancer cell models. In addition to the in vivo findings, IL-1 contributed to the production of vascular endothelial cell growth factor and tumor necrosis factor in cocultures of peritoneal macrophages and tumor cells. Host-derived IL-1 seems to control tumor angiogenesis and invasiveness. Furthermore, the anti-angiogenic effects of IL-1 receptor antagonist, shown here, suggest a possible therapeutic role in cancer, in addition to its current use in rheumatoid arthritis.


Journal of Immunology | 2011

IL-1α and IL-1β Recruit Different Myeloid Cells and Promote Different Stages of Sterile Inflammation

Peleg Rider; Yaron Carmi; Ofer Guttman; Alex Braiman; Idan Cohen; Elena Voronov; Malka R. White; Charles A. Dinarello; Ron N. Apte

The immune system has evolved to protect the host from invading pathogens and to maintain tissue homeostasis. Although the inflammatory process involving pathogens is well documented, the intrinsic compounds that initiate sterile inflammation and how its progression is mediated are still not clear. Because tissue injury is usually associated with ischemia and the accompanied hypoxia, the microenvironment of various pathologies involves anaerobic metabolites and products of necrotic cells. In the current study, we assessed in a comparative manner the role of IL-1α and IL-1β in the initiation and propagation of sterile inflammation induced by products of hypoxic cells. We found that following hypoxia, the precursor form of IL-1α, and not IL-1β, is upregulated and subsequently released from dying cells. Using an inflammation-monitoring system consisting of Matrigel mixed with supernatants of hypoxic cells, we noted accumulation of IL-1α in the initial phase, which correlated with the infiltration of neutrophils, and the expression of IL-1β correlated with later migration of macrophages. In addition, we were able to show that IL-1 molecules from cells transfected with either precursor IL-1α or mature IL-1β can recruit neutrophils or macrophages, respectively. Taken together, these data suggest that IL-1α, released from dying cells, initiates sterile inflammation by inducing recruitment of neutrophils, whereas IL-1β promotes the recruitment and retention of macrophages. Overall, our data provide new insight into the biology of IL-1 molecules as well as on the regulation of sterile inflammation.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Differential release of chromatin-bound IL-1α discriminates between necrotic and apoptotic cell death by the ability to induce sterile inflammation

Idan Cohen; Peleg Rider; Yaron Carmi; Alex Braiman; Shahar Dotan; Malka R. White; Elena Voronov; Michael Martin; Charles A. Dinarello; Ron N. Apte

IL-1α, like IL-1β, possesses multiple inflammatory and immune properties. However, unlike IL-1β, the cytokine is present intracellularly in healthy tissues and is not actively secreted. Rather, IL-1α translocates to the nucleus and participates in transcription. Here we show that intracellular IL-1α is a chromatin-associated cytokine and highly dynamic in the nucleus of living cells. During apoptosis, IL-1α concentrates in dense nuclear foci, which markedly reduces its mobile nature. In apoptotic cells, IL-1α is retained within the chromatin fraction and is not released along with the cytoplasmic contents. To simulate the in vivo inflammatory response to cells undergoing different mechanisms of death, lysates of cells were embedded in Matrigel plugs and implanted into mice. Lysates from cells undergoing necrosis recruited cells of the myeloid lineage into the Matrigel, whereas lysates of necrotic cells lacking IL-1α failed to recruit an infiltrate. In contrast, lysates of cells undergoing apoptotic death were inactive. Cells infiltrating the Matrigel were due to low concentrations (20–50 pg) of the IL-1α precursor containing the receptor interacting C-terminal, whereas the N-terminal propiece containing the nuclear localization site failed to do so. When normal keratinocytes were subjected to hypoxia, the constitutive IL-1α precursor was released into the supernatant. Thus, after an ischemic event, the IL-1α precursor is released by hypoxic cells and incites an inflammatory response by recruiting myeloid cells into the area. Tissues surrounding the necrotic site also sustain damage from the myeloid cells. Nuclear trafficking and differential release during necrosis vs. apoptosis demonstrate that inflammation by IL-1α is tightly controlled.


Journal of Immunology | 2005

CD11b+/Gr-1+ Immature Myeloid Cells Mediate Suppression of T Cells in Mice Bearing Tumors of IL-1β-Secreting Cells

Xiaoping Song; Yakov Krelin; Tatyana Dvorkin; Olle Bjorkdahl; Shraga Segal; Charles A. Dinarello; Elena Voronov; Ron N. Apte

Tumor cells secreting IL-1β are invasive and metastatic, more than the parental line or control mock-transfected cells, and concomitantly induce in mice general immune suppression of T cell responses. Suppression strongly correlates with accumulation in the peripheral blood and spleen of CD11b+/Gr-1+ immature myeloid cells and hematological alterations, such as splenomegaly, leukocytosis, and anemia. Resection of large tumors of IL-1β-secreting cells restored immune reactivity and hematological alterations within 7–10 days. Treatment of tumor-bearing mice with the physiological inhibitor of IL-1, the IL-1R antagonist, reduced tumor growth and attenuated the hematological alterations. Depletion of CD11b+/Gr-1+ immature myeloid cells from splenocytes of tumor-bearing mice abrogated suppression. Despite tumor-mediated suppression, resection of large tumors of IL-1β-secreting cells, followed by a challenge with the wild-type parental cells, induced resistance in mice; protection was not observed in mice bearing tumors of mock-transfected fibrosarcoma cells. Altogether, we show in this study that tumor-derived IL-1β, in addition to its proinflammatory effects on tumor invasiveness, induces in the host hematological alterations and tumor-mediated suppression. Furthermore, the antitumor effectiveness of the IL-1R antagonist was also shown to encompass restoration of hematological alterations, in addition to its favorable effects on tumor invasiveness and angiogenesis that have previously been described by us.


Fertility and Sterility | 1991

Cytokine involvement in oocytes and early embryos

Mati Zolti; Zion Ben-Rafael; Ruth Meirom; Mordechai Shemesh; David Bider; Shlomo Mashiach; Ron N. Apte

OBJECTIVE The early events of reproduction involve a carefully modulated complex system of oocyte maturation, fertilization, and proliferation. The aim of the study was to measure the presence of cytokines, namely interleukin 1 (IL-1), interleukin 6 (IL-6), colony-stimulating factor 1 (CSF-1), and tumor necrosis factor (TNF) in the conditioned medium (CM) of the oocytes, granulosa cells, cumulus cells, one to eight-cell embryos and sperm. DESIGN The material was obtained from men and women undergoing in vitro fertilization therapy. MAIN OUTCOME MEASURES We hypothesized that cytokines might affect embryonic growth and differentiation as they show a pleotropic effect on immune cells. RESULTS All these cytokines are present in significant quantities in the CM and were shown to be expressed in a sequential manner; thus, some are present in the oocyte and its vestment, the corona-cumulus complex (IL-1, IL-6, and CSF-1), whereas TNF appears only at the stage of six to eight-cell embryos. Inflammatory cytokines could not be detected in sperm samples. CONCLUSIONS It is possible that these cytokines have a role in the regulation of embryonic development, maternal immunological recognition of pregnancy, and maintenance of proper hormonal environment.


Cancer Research | 2007

Interleukin-1β–Driven Inflammation Promotes the Development and Invasiveness of Chemical Carcinogen–Induced Tumors

Yakov Krelin; Elena Voronov; Shahar Dotan; Moshe Elkabets; Eli Reich; Mina Fogel; Monika Huszar; Yoichiro Iwakura; Shraga Segal; Charles A. Dinarello; Ron N. Apte

The role of microenvironment interleukin 1 (IL-1) on 3-methylcholanthrene (3-MCA)-induced carcinogenesis was assessed in IL-1-deficient mice, i.e., IL-1beta(-/-), IL-1alpha(-/-), IL-1alpha/beta(-/-) (double knockout), and mice deficient in the naturally occurring inhibitor of IL-1, the IL-1 receptor antagonist (IL-1Ra). Tumors developed in all wild-type (WT) mice, whereas in IL-1beta-deficient mice, tumors developed slower and only in some of the mice. In IL-1Ra-deficient mice, tumor development was the most rapid. Tumor incidence was similar in WT and IL-1alpha-deficient mice. Histologic analyses revealed fibrotic structures forming a capsule surrounding droplets of the carcinogen in olive oil, resembling foreign body-like granulomas, which appeared 10 days after injection of 3-MCA and persisted until the development of local tumors. A sparse leukocyte infiltrate was found at the site of carcinogen injection in IL-1beta-deficient mice, whereas in IL-1Ra-deficient mice, a dense neutrophilic infiltrate was observed. Treatment of IL-1Ra-deficient mice with recombinant IL-1Ra but not with an inhibitor of tumor necrosis factor abrogated the early leukocytic infiltrate. The late leukocyte infiltrate (day 70), which was dominated by macrophages, was also apparent in WT and IL-1alpha-deficient mice, but was nearly absent in IL-1beta-deficient mice. Fibrosarcoma cell lines, established from 3-MCA-induced tumors from IL-1Ra-deficient mice, were more aggressive and metastatic than lines from WT mice; cell lines from IL-1-deficient mice were the least invasive. These observations show the crucial role of microenvironment-derived IL-1beta, rather than IL-1alpha, in chemical carcinogenesis and in determining the invasive potential of malignant cells.


Seminars in Cancer Biology | 2002

Interleukin-1—a major pleiotropic cytokine in tumor–host interactions

Ron N. Apte; Elena Voronov

Interleukin-1 (IL-1) represents a family of two agonistic proteins, IL-1alpha and IL-1beta, that are pleiotropic and affect hemopoiesis, inflammation, and immunity. In the context of the producing cell, IL-1beta is solely active in its secreted form, whereas IL-1alpha is active as an intracellular precursor, as a membrane-associated cytokine and to a lesser extent as a secreted molecule. IL-1 is abundant at tumor sites, where it may not only affect the growth and invasiveness of malignant cells, but where it may also induce antitumor immunity. Here we review the effects of microenvironmental and tumor cell-associated IL-1 on malignant processes, in experimental tumor models and in cancer patients.


Immunological Reviews | 2008

Is interleukin-1 a good or bad ‘guy’ in tumor immunobiology and immunotherapy?

Ron N. Apte; Elena Voronov

Summary: The interleukin‐1 (IL‐1) family consists of two major agonistic proteins, IL‐1α and IL‐1β, which are pleiotropic and affect mainly inflammation, immunity, and hemopoiesis. The IL‐1 receptor antagonist (IL‐1Ra) is a physiological inhibitor of pre‐formed IL‐1. In their secreted form, IL‐1α and IL‐1β bind to the same receptors and induce the same biological functions. However, the IL‐1 molecules differ in their compartmentalization within the producing cell or the microenvironment. Thus, IL‐1β is solely active in its secreted form, whereas IL‐1α is mainly active in cell‐associated forms (intracellular precursor and membrane‐bound IL‐1) and only rarely as a secreted cytokine, mainly by macrophages/monocytes. IL‐1 is abundant at tumor sites, being produced by cellular elements of the tumor microenvironment or by the malignant cells, and it affects not only various phases of the malignant process, such as carcinogenesis, tumor growth, and invasiveness, but also patterns of interactions between malignant cells and the hosts immune system. Hence, the effects of the IL‐1 molecules on the malignant process are complex and are often of an opposing nature. Comparative studies on the differential roles of malignant cell‐ or host‐derived IL‐1α and IL‐1β in different stages of the malignant process can subsequently open new avenues for manipulation of IL‐1 expression and function in cancer immunotherapy.


Journal of Immunology | 2009

The Role of Macrophage-Derived IL-1 in Induction and Maintenance of Angiogenesis

Yaron Carmi; Elena Voronov; Shahar Dotan; Nitza Lahat; Michal A. Rahat; Mina Fogel; Monika Huszar; Malka R. White; Charles A. Dinarello; Ron N. Apte

Inflammation and angiogenesis are pivotal processes in the progression of many diseases, including malignancies. A hypoxic microenvironment often results in a milieu of proinflammatory and proangiogenic cytokines produced by infiltrating cells. We assessed the role of macrophage-derived hypoxia-associated cytokines in promoting inflammation and angiogenesis. Supernatants of macrophages, stimulated under hypoxia with or without an inflammatory stimulus (LPS), promoted angiogenesis when incorporated into Matrigel plugs. However, neutralization of IL-1 in the supernatants, particularly IL-1β, completely abrogated cell infiltration and angiogenesis in Matrigel plugs and reduced vascular endothelial growth factor (VEGF) levels by 85%. Similarly, supernatants from macrophages of IL-1β knockout mice did not induce inflammatory or angiogenic responses. The importance of IL-1 signaling in the host was demonstrated by the dramatic reduction of inflammatory and angiogenic responses in Matrigel plugs that contained macrophage supernatants from control mice which had been implanted in IL-1 receptor type I knockout mice. Myeloid cells infiltrating into Matrigel plugs were of bone marrow origin and represented the major source of IL-1 and other cytokines/chemokines in the plugs. Cells of endothelial lineage were the main source of VEGF and were recruited mainly from neighboring tissues, rather than from the bone marrow. Using the aortic ring sprouting assay, it was shown that in this experimental system, IL-1 does not directly activate endothelial cell migration, proliferation and organization into blood vessel-like structures, but rather activates infiltrating cells to produce endothelial cell activating factors, such as VEGF. Thus, targeting IL-1β has the potential to inhibit angiogenesis in pathological situations and may be of considerable clinical value.


European Journal of Immunology | 2010

IL-1β regulates a novel myeloid-derived suppressor cell subset that impairs NK cell development and function.

Moshe Elkabets; Vera S. G. Ribeiro; Charles A. Dinarello; Suzanne Ostrand-Rosenberg; James P. Di Santo; Ron N. Apte; Christian A. J. Vosshenrich

Chronic inflammation is associated with promotion of malignancy and tumor progression. Many tumors enhance the accumulation of myeloid‐derived suppressor cells (MDSC), which contribute to tumor progression and growth by suppressing anti‐tumor immune responses. Tumor‐derived IL‐1β secreted into the tumor microenvironment has been shown to induce the accumulation of MDSC possessing an enhanced capacity to suppress T cells. In this study, we found that the enhanced suppressive potential of IL‐1β‐induced MDSC was due to the activity of a novel subset of MDSC lacking Ly6C expression. This subset was present at low frequency in tumor‐bearing mice in the absence of IL‐1β‐induced inflammation; however, under inflammatory conditions, Ly6Cneg MDSC were predominant. Ly6Cneg MDSC impaired NK cell development and functions in vitro and in vivo. These results identify a novel IL‐1β‐induced subset of MDSC with unique functional properties. Ly6Cneg MDSC mediating NK cell suppression may thus represent useful targets for therapeutic interventions.

Collaboration


Dive into the Ron N. Apte's collaboration.

Top Co-Authors

Avatar

Elena Voronov

Ben-Gurion University of the Negev

View shared research outputs
Top Co-Authors

Avatar

Shraga Segal

Ben-Gurion University of the Negev

View shared research outputs
Top Co-Authors

Avatar

Charles A. Dinarello

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Shahar Dotan

Ben-Gurion University of the Negev

View shared research outputs
Top Co-Authors

Avatar

Yaron Carmi

Ben-Gurion University of the Negev

View shared research outputs
Top Co-Authors

Avatar

Moshe Elkabets

Ben-Gurion University of the Negev

View shared research outputs
Top Co-Authors

Avatar

Rosalyn M. White

Ben-Gurion University of the Negev

View shared research outputs
Top Co-Authors

Avatar

Margot Zöller

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Tatyana Dvorkin

Ben-Gurion University of the Negev

View shared research outputs
Top Co-Authors

Avatar

Peleg Rider

Ben-Gurion University of the Negev

View shared research outputs
Researchain Logo
Decentralizing Knowledge