Network


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

Hotspot


Dive into the research topics where Anna Kajaste-Rudnitski is active.

Publication


Featured researches published by Anna Kajaste-Rudnitski.


Journal of Biological Chemistry | 2006

The 2′,5′-Oligoadenylate Synthetase 1b Is a Potent Inhibitor of West Nile Virus Replication Inside Infected Cells

Anna Kajaste-Rudnitski; Tomoji Mashimo; Marie Pascale Frenkiel; Jean Louis Guénet; Marianne Lucas; Philippe Desprès

The 2′,5′-oligoadenylate synthetase (OAS) proteins associated with endoribonuclease RNase L are components of the interferon-regulated OAS/RNase L system, which is an RNA decay pathway known to play an important role in the innate antiviral immunity. A large body of evidence suggests a critical role for the 1b isoform of the mouse Oas gene (Oas1b) in resistance to West Nile virus (WNV) infection in vivo. WNV is a positive, single-stranded RNA virus responsible for severe encephalitis in a large range of animal species and humans. To investigate the molecular basis for the sensitivity of WNV to the Oas1b antiviral pathway, we established a stable mouse fibroblastic cell clone that up-regulates Oas1b protein expression under the control of the Tet-Off expression system. We showed that murine cells respond to Oas1b expression by efficiently inhibiting WNV replication. The antiviral action of Oas1b was essentially restricted to the early stages in virus life cycle. We found that the inability of WNV to productively infect the Oas1b-expressing cells was attributable to a dramatic reduction in positive-stranded viral RNA level. Thus, Oas1b represents an antiviral pathway that exerts its inhibitory effect on WNV replication by preventing viral RNA accumulation inside infected cells.


Journal of Virology | 2013

TRIM22 Inhibits Influenza A Virus Infection by Targeting the Viral Nucleoprotein for Degradation

Andrea Di Pietro; Anna Kajaste-Rudnitski; Alexandra Oteiza; Lucia Nicora; Greg J. Towers; Nadir Mechti; Elisa Vicenzi

ABSTRACT Tripartite motif (TRIM) protein superfamily members are emerging as important effectors of the innate immune response against viral infections. In particular, TRIM22 was reported to exert antiviral activity against RNA viruses, such as hepatitis B virus (HBV), encephalomyocarditis virus (ECMV), and human immunodeficiency virus type 1 (HIV-1). We demonstrate here, for the first time, that TRIM22 is upregulated by influenza A virus (IAV) infection at both mRNA and protein levels in human alveolar epithelial A549 cells. Conversely, TRIM22 potently restricted IAV replication, in that prevention of TRIM22 expression by means of short hairpin RNA led to a 10-fold enhancement of IAV replication in these cells. Depletion of TRIM22 also reduced the anti-IAV activity of alpha interferon (IFN-α), suggesting that TRIM22 is an important IFN-stimulated gene that is required for maximal suppression of IAV by type I IFN. Furthermore, the IAV infectious titer decreased up to 100-fold in MDCK cells expressing exogenous human TRIM22. Restriction of IAV replication was accounted for by the interaction between TRIM22 and the viral nucleoprotein (NP), resulting in its polyubiquitination and degradation in a proteasome-dependent manner. Thus, TRIM22 represents a novel restriction factor upregulated upon IAV infection that curtails its replicative capacity in epithelial cells.


Journal of Virology | 2011

TRIM22 Inhibits HIV-1 Transcription Independently of Its E3 Ubiquitin Ligase Activity, Tat, and NF-κB-Responsive Long Terminal Repeat Elements

Anna Kajaste-Rudnitski; Sara S. Marelli; Cinzia Pultrone; Thomas Pertel; Pradeep D. Uchil; Nadir Mechti; Walther Mothes; Guido Poli; Jeremy Luban; Elisa Vicenzi

ABSTRACT Previous studies identified clones of the U937 promonocytic cell line that were either permissive or nonpermissive for human immunodeficiency virus type 1 (HIV-1) replication. These clones were investigated further in the search for host restriction factors that could explain their differential capacity to support HIV-1 replication. Among known HIV-1 restriction factors screened, tripartite motif-containing protein 22 (TRIM22) was the only factor constitutively expressed in nonpermissive and absent in permissive U937 cells. Stable TRIM22 knockdown (KD) rescued HIV-1 long-terminal-repeat (LTR)-driven transcription in KD-nonpermissive cells to the levels observed in permissive cells. Conversely, transduction-mediated expression of TRIM22 in permissive cells reduced LTR-driven luciferase expression by ∼7-fold, supporting a negative role of TRIM22 in HIV-1 transcription. This finding was further confirmed in the human T cell line A3.01 expressing TRIM22. Moreover, overexpression of TRIM22 in 293T cells significantly impaired basal and phorbol myristate acetate-ionomycin-induced HIV-1 LTR-driven gene expression, whereas inhibition of tumor necrosis factor alpha-induced viral transcription was a consequence of lower basal expression. In agreement, TRIM22 equally inhibited an LTR construct lacking the tandem NF-κB binding sites. In addition, TRIM22 did not affect Tat-mediated LTR transactivation. Finally, these effects were independent of TRIM22 E3 ubiquitin-ligase activity. In the context of replication-competent virus, significantly higher levels of HIV-1 production were observed in KD-nonpermissive versus control nonpermissive U937 cells after infection. In contrast, lower peak levels of HIV-1 replication characterized U937 and A3.01 cells expressing TRIM22 versus their control transduced counterpart. Thus, nuclear TRIM22 significantly impairs HIV-1 replication, likely by interfering with Tat- and NF-κB-independent LTR-driven transcription.


Journal of Virology | 2011

TRIM22 Inhibits HIV-1 Transcription Independently of Its E3-Ubiquitin Ligase Activity, Tat and NF-κB Responsive LTR Elements

Anna Kajaste-Rudnitski; Sara S. Marelli; Cinzia Pultrone; Thomas Pertel; Pradeep D. Uchil; Nadir Mechti; Walther Mothes; Guido Poli; Jeremy Luban; Elisa Vicenzi

ABSTRACT Previous studies identified clones of the U937 promonocytic cell line that were either permissive or nonpermissive for human immunodeficiency virus type 1 (HIV-1) replication. These clones were investigated further in the search for host restriction factors that could explain their differential capacity to support HIV-1 replication. Among known HIV-1 restriction factors screened, tripartite motif-containing protein 22 (TRIM22) was the only factor constitutively expressed in nonpermissive and absent in permissive U937 cells. Stable TRIM22 knockdown (KD) rescued HIV-1 long-terminal-repeat (LTR)-driven transcription in KD-nonpermissive cells to the levels observed in permissive cells. Conversely, transduction-mediated expression of TRIM22 in permissive cells reduced LTR-driven luciferase expression by ∼7-fold, supporting a negative role of TRIM22 in HIV-1 transcription. This finding was further confirmed in the human T cell line A3.01 expressing TRIM22. Moreover, overexpression of TRIM22 in 293T cells significantly impaired basal and phorbol myristate acetate-ionomycin-induced HIV-1 LTR-driven gene expression, whereas inhibition of tumor necrosis factor alpha-induced viral transcription was a consequence of lower basal expression. In agreement, TRIM22 equally inhibited an LTR construct lacking the tandem NF-κB binding sites. In addition, TRIM22 did not affect Tat-mediated LTR transactivation. Finally, these effects were independent of TRIM22 E3 ubiquitin-ligase activity. In the context of replication-competent virus, significantly higher levels of HIV-1 production were observed in KD-nonpermissive versus control nonpermissive U937 cells after infection. In contrast, lower peak levels of HIV-1 replication characterized U937 and A3.01 cells expressing TRIM22 versus their control transduced counterpart. Thus, nuclear TRIM22 significantly impairs HIV-1 replication, likely by interfering with Tat- and NF-κB-independent LTR-driven transcription.


Science Translational Medicine | 2014

Genetic engineering of hematopoiesis for targeted IFN-α delivery inhibits breast cancer progression.

Giulia Escobar; Davide Moi; Anna Ranghetti; Pinar Ozkal-Baydin; Mario Leonardo Squadrito; Anna Kajaste-Rudnitski; Attilio Bondanza; Bernhard Gentner; Michele De Palma; Roberta Mazzieri; Luigi Naldini

Inhibition of primary and metastatic breast cancer by targeted delivery of interferon-α by tumor-infiltrating monocytes. Toasting Tumor Immunotherapy Tumor immunotherapy is a promising new strategy for cancer treatment but is somewhat limited by the immunosuppressive nature of the tumor microenvironment. Type I interferons (IFNs) have been shown to promote tumor immunity, but systemic toxicity has limited their use. Genetic engineering of hematopoietic stem cells (HSCs) is one way to modulate the immune response in the tumor microenvironment. Now, Escobar et al. attempt to overcome this immunosuppression by introducing IFN-α into tumor-infiltrating macrophages. The authors developed a way to insert an IFN-α transgene into HSCs, but restrict IFN-α expression to differentiated monocytes. They then tested their cells in human hematochimeric mice. HSC engraftment and repopulation is inhibited if IFN-α is expressed in the less differentiated cells. However, here, the HSCs engraft and repopulate NSG mice. What’s more, these cells inhibit tumor progression and experimental metastasis in an autologous model of breast cancer, effectively reprogramming the tumor microenvironment. The immunosuppressive tumor microenvironment represents a major hurdle to cancer therapy. We developed a gene transfer strategy into hematopoietic stem cells (HSCs) to target transgene expression to tumor-infiltrating monocytes/macrophages. Using a combination of transcriptional and microRNA-mediated control, we achieved selective expression of an interferon-α (IFN-α) transgene in differentiated monocytes of human hematochimeric mice. We show that IFN-α transgene expression does not impair engraftment and long-term multilineage repopulation of NSG (NOD/LtSz-scidIL2Rγnull) mice by transplanted human HSCs. By providing a source of human cytokines in the mice, we improved the functional reconstitution of human myeloid, natural killer, and T cell lineages, and achieved enhanced immune-mediated clearance of transplanted human breast tumors when hematopoiesis was engineered for tumor-targeted IFN-α expression. By applying our strategy to mouse breast cancer models, we achieved inhibition of tumor progression and experimental metastases in an autologous setting, likely through enhanced generation of effector T cells and their recruitment to the neoplastic tissues. By forcing IFN-α expression in tumor-infiltrating macrophages, we blunted their innate protumoral activity and reprogrammed the tumor microenvironment toward more effective dendritic cell activation and immune effector cell cytotoxicity. Overall, our studies validate the feasibility, safety, and therapeutic potential of a new cancer gene therapy strategy, and open the way to test this approach as adjuvant therapy in advanced breast cancer patients.


AIDS | 2011

Induction of protective antibody response by MF59-adjuvanted 2009 pandemic A/H1N1v influenza vaccine in HIV-1-infected individuals

Anna Kajaste-Rudnitski; Laura Galli; Silvia Nozza; Giuseppe Tambussi; Andrea Di Pietro; Gabriele Pellicciotta; Alessandro Monti; Paolo Mascagni; Matteo Moro; Elisa Vicenzi

Objective:To determine the immunogenicity of the monovalent vaccine against 2009 pandemic influenza A/H1N1 in HIV-1-infected individuals. Design:A total of 192 participants, including 44 HIV-1-positive individuals and 148 HIV-1-negative healthy controls were enrolled to receive a single dose of MF59-adjuvanted 2009 A/H1N1v vaccine formulated to contain 7.5 μg of haemagglutin antigen. Methods:Standard haemagglutination inhibition (HAI) assay was performed to evaluate seroconversion and seroprotecsion rates against the pandemic virus in serum samples collected at baseline (T0) and 3–5-week postvaccination (T28). Seroconversion to vaccination was defined by either prevaccination HAI titer less than 1: 10 with a postvaccination titer higher than 1: 40, or a prevaccination titer higher than 1: 10 and increase of at least four-fold or more after vaccination. Seroprotection was defined by HAI titers higher than 1: 40. Results:The vaccine induced specific antibody titers in HIV-1-positive individuals similar to those of HIV-1-negative controls [215.3, 95% confidence interval (CI) 150.4–308.1 vs. 275.9, 95% CI 232.6–327.3] with postvaccination seroprotection rates higher than 97%. In contrast, the seroconversion rate was lower in the HIV-1-positive individuals as compared with the HIV-1-negative controls (36.4 vs. 79.0%, P < 0.0001), likely as a consequence of their high HAI baseline titers. Multivariable logistic regression analysis showed that seroconversion was less likely in HIV-1-positive individuals [odds ratio (OR) = 0.237, 95% CI 0.104–0.539, P = 0.0006) and with increasing age (OR = 0.805, 95% CI 0.684–0.947, P = 0.009). Conclusions:A single dose of MF59-adjuvanted 2009 influenza H1N1 vaccine induced an immune response against pandemic H1N1 virus in HIV-1-positive individuals reaching titers similar to those of HIV-1-negative individuals. The seroconversion rate was negatively associated with HIV infection and increasing age.


Molecular Therapy | 2015

Dual-regulated Lentiviral Vector for Gene Therapy of X-linked Chronic Granulomatosis

Maria Chiriaco; Giada Farinelli; Valentina Capo; Erika Zonari; Samantha Scaramuzza; Gigliola Di Matteo; Lucia Sergi Sergi; Maddalena Migliavacca; Raisa Jofra Hernandez; Ferdinando Bombelli; Ezio Giorda; Anna Kajaste-Rudnitski; Didier Trono; Manuel Grez; Paolo Rossi; Andrea Finocchi; Luigi Naldini; Bernhard Gentner; Alessandro Aiuti

Regulated transgene expression may improve the safety and efficacy of hematopoietic stem cell (HSC) gene therapy. Clinical trials for X-linked chronic granulomatous disease (X-CGD) employing gammaretroviral vectors were limited by insertional oncogenesis or lack of persistent engraftment. Our novel strategy, based on regulated lentiviral vectors (LV), targets gp91(phox) expression to the differentiated myeloid compartment while sparing HSC, to reduce the risk of genotoxicity and potential perturbation of reactive oxygen species levels. Targeting was obtained by a myeloid-specific promoter (MSP) and posttranscriptional, microRNA-mediated regulation. We optimized both components in human bone marrow (BM) HSC and their differentiated progeny in vitro and in a xenotransplantation model, and generated therapeutic gp91(phox) expressing LVs for CGD gene therapy. All vectors restored gp91(phox) expression and function in human X-CGD myeloid cell lines, primary monocytes, and differentiated myeloid cells. While unregulated LVs ectopically expressed gp91(phox) in CD34(+) cells, transcriptionally and posttranscriptionally regulated LVs substantially reduced this off-target expression. X-CGD mice transplanted with transduced HSC restored gp91(phox) expression, and MSP-driven vectors maintained regulation during BM development. Combining transcriptional (SP146.gp91-driven) and posttranscriptional (miR-126-restricted) targeting, we achieved high levels of myeloid-specific transgene expression, entirely sparing the CD34(+) HSC compartment. This dual-targeted LV construct represents a promising candidate for further clinical development.


AIDS | 2013

M1 polarization of human monocyte-derived macrophages restricts pre and postintegration steps of HIV-1 replication.

Luca Cassetta; Anna Kajaste-Rudnitski; Tiziana Coradin; Elisa Saba; Giulia Della Chiara; Marialuisa Barbagallo; Francesca Graziano; Massimo Alfano; Edana Cassol; Elisa Vicenzi; Guido Poli

Objective:Functional polarization of human monocyte-derived macrophages (MDMs) into M1 cells leads to inhibition of R5 HIV-1 replication and viral DNA synthesis in comparison to control, unpolarized cells together with CD4 downregulation from the cell surface and upregulation of CCR5-binding chemokine secretion. We here investigated whether a postentry restriction of virus replication is also induced by M1 polarization of MDM. Design:MDM were first polarized to M1 cells by 18 h stimulation with interferon-&ggr; and tumor necrosis factor-&agr;; the cytokines were then removed and the cells were infected with vesicular stomatitis virus G-protein pseudotyped enhanced green fluorescence protein HIV-1 (HIV-GFP) generating a single-round infection cycle. Methods:HIV-1 expression was monitored in terms of eGFP expression by fluorescence activated cell sorter (FACS) analysis and real-time PCR analysis of total HIV-1 gag DNA, 2-long terminal repeat DNA, proviral DNA, and multiply spliced RNA transcripts. Expression of apolipopoprotein B mRNA-editing, enzyme-catalytic, polypeptide-like 3G (APOBEC3G), and APOBEC3A was tested by western blotting and FACS analysis. Results:Inhibition of HIV-GFP expression was observed in M1-MDM along with impaired viral DNA synthesis, delayed proviral integration, and reduced proviral transcription. Although APOBEC3G levels were similar in M1 and unpolarized MDM, APOBEC 3A was selectively expressed only by M1 cells. Conclusion:M1 polarization of in-vitro differentiated primary MDM determines a transient, but profound restriction of HIV-1 replication affecting multiple (entry and postentry) steps in the virus life cycle likely involving the upregulated expression of APOBEC3A.


Stem cell reports | 2017

Efficient Ex Vivo Engineering and Expansion of Highly Purified Human Hematopoietic Stem and Progenitor Cell Populations for Gene Therapy

Erika Zonari; Giacomo Desantis; Carolina Petrillo; Francesco Boccalatte; Maria Rosa Lidonnici; Anna Kajaste-Rudnitski; Alessandro Aiuti; Giuliana Ferrari; Luigi Naldini; Bernhard Gentner

Summary Ex vivo gene therapy based on CD34+ hematopoietic stem cells (HSCs) has shown promising results in clinical trials, but genetic engineering to high levels and in large scale remains challenging. We devised a sorting strategy that captures more than 90% of HSC activity in less than 10% of mobilized peripheral blood (mPB) CD34+ cells, and modeled a transplantation protocol based on highly purified, genetically engineered HSCs co-infused with uncultured progenitor cells. Prostaglandin E2 stimulation allowed near-complete transduction of HSCs with lentiviral vectors during a culture time of less than 38 hr, mitigating the negative impact of standard culture on progenitor cell function. Exploiting the pyrimidoindole derivative UM171, we show that transduced mPB CD34+CD38− cells with repopulating potential could be expanded ex vivo. Implementing these findings in clinical gene therapy protocols will improve the efficacy, safety, and sustainability of gene therapy and generate new opportunities in the field of gene editing.


Antimicrobial Agents and Chemotherapy | 2008

Inhibition of Herpes Simplex Virus Types 1 and 2 In Vitro Infection by Sulfated Derivatives of Escherichia coli K5 Polysaccharide

Debora Pinna; Pasqua Oreste; Tiziana Coradin; Anna Kajaste-Rudnitski; Silvia Ghezzi; Giorgio Zoppetti; Antonella Rotola; Rafaela Argnani; Guido Poli; Roberto Manservigi; Elisa Vicenzi

ABSTRACT Herpes simplex virus type 1 (HSV-1) and HSV-2 are neurotropic viruses and common human pathogens causing major public health problems such as genital herpes, a sexually transmitted disease also correlated with increased transmission and replication of human immunodeficiency virus type 1 (HIV-1). Therefore, compounds capable of blocking HIV-1, HSV-1, and HSV-2 transmission represent candidate microbicides with a potential added value over that of molecules acting selectively against either infection. We report here that sulfated derivatives of the Escherichia coli K5 polysaccharide, structurally highly similar to heparin and previously shown to inhibit HIV-1 entry and replication in vitro, also exert suppressive activities against both HSV-1 and HSV-2 infections. In particular, the N,O-sulfated [K5-N,OS(H)] and O-sulfated epimerized [Epi-K5-OS(H)] forms inhibited the infection of Vero cells by HSV-1 and -2, with 50% inhibitory concentrations (IC50) between 3 ± 0.05 and 48 ± 27 nM, and were not toxic to the cells at concentrations as high as 5 μM. These compounds impaired the early steps of HSV-1 and HSV-2 virion attachment and entry into host cells and reduced the cell-to-cell spread of HSV-2. Since K5-N,OS(H) and Epi-K5-OS(H) also inhibit HIV-1 infection, they may represent valid candidates for development as topical microbicides preventing sexual transmission of HIV-1, HSV-1, and HSV-2.

Collaboration


Dive into the Anna Kajaste-Rudnitski's collaboration.

Top Co-Authors

Avatar

Elisa Vicenzi

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar

Guido Poli

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar

Luigi Naldini

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar

Bernhard Gentner

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar

Alessandro Aiuti

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar

Carolina Petrillo

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar

Silvia Ghezzi

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar

Jeremy Luban

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nadir Mechti

Centre national de la recherche scientifique

View shared research outputs
Researchain Logo
Decentralizing Knowledge