2,037 results on '"CD46"'
Search Results
152. Compound Haplotype Variants in CFH and CD46 Genes Determine Clinical Outcome of Atypical Hemolytic Uremic Syndrome (aHUS)—A Series of Cases from a Single Family
- Author
-
Monika Kolanowska, Krystian Jazdzewski, Marta Kotlarek-Łysakowska, Magdalena Durlik, Anna Kubiak-Dydo, Katarzyna Salata, Agnieszka Furmańczyk-Zawiska, Pawel Gaj, Michał Świerniak, Anna Wojcicka, Ewelina Użarowska-Gąska, Beata Leszczyńska, and Maria Daniel
- Subjects
Hemolytic anemia ,030232 urology & nephrology ,lcsh:Medicine ,Medicine (miscellaneous) ,kidney transplantation ,Case Report ,urologic and male genital diseases ,03 medical and health sciences ,aHUS ,0302 clinical medicine ,hemic and lymphatic diseases ,Atypical hemolytic uremic syndrome ,Medicine ,CD46 ,Kidney transplantation ,030304 developmental biology ,0303 health sciences ,business.industry ,lcsh:R ,Eculizumab ,medicine.disease ,Penetrance ,Factor H ,Immunology ,CFH ,hemolytic-uremic syndrome ,business ,medicine.drug ,Rare disease - Abstract
Atypical hemolytic uremic syndrome (aHUS) is a rare disease triggered by dysregulation of the alternative complement pathway, consisting of a characteristic triad of nonimmune hemolytic anemia, thrombocytopenia, and renal failure. The risk of aHUS onset, recurrence, and allograft loss depends on the genetic background of a patient. We show a series of cases from a single family whose five members were affected by aHUS and presented distinct clinical outcomes. Next-generation sequencing revealed combined mutations in both complement factor H and membrane cofactor protein CD46. Out of eight siblings, aHUS affected three adult brothers, and, subsequently, affected two children of an unaffected sister. The first patient died due to aHUS, and two other brothers underwent successful kidney transplantation with no aHUS recurrence. The younger, 10-month-old child presented with a severe course of the disease with cardiac involvement and persistent hemolytic anemia limited by eculizumab, while the 2-year-old recovered completely on eculizumab. The study shows a highly variable disease penetrance.
- Published
- 2021
153. Microglial trogocytosis and the complement system regulate axonal pruning in vivo
- Author
-
Tony KY Lim and Edward S Ruthazer
- Subjects
Retinal Ganglion Cells ,0301 basic medicine ,Xenopus ,pruning ,microglia ,Cell Communication ,Xenopus Proteins ,Xenopus laevis ,Immunology and Inflammation ,0302 clinical medicine ,Biology (General) ,Axon ,Complement Activation ,axon ,Neuronal Plasticity ,Microglia ,General Neuroscience ,General Medicine ,Cell biology ,medicine.anatomical_structure ,Retinal ganglion cell ,Medicine ,morphometry ,Research Article ,Trogocytosis ,QH301-705.5 ,Science ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Membrane Cofactor Protein ,03 medical and health sciences ,Phagocytosis ,medicine ,Animals ,Humans ,General Immunology and Microbiology ,behavior ,CD46 ,biology.organism_classification ,Axons ,Complement system ,030104 developmental biology ,nervous system ,Synapses ,retinotectal ,030217 neurology & neurosurgery ,Ex vivo ,Neuroscience - Abstract
Partial phagocytosis—called trogocytosis—of axons by microglia has been documented in ex vivo preparations but has not been directly observed in vivo. The mechanisms that modulate microglial trogocytosis of axons and its function in neural circuit development remain poorly understood. Here we directly observe axon trogocytosis by microglia in vivo in the developing Xenopus laevis retinotectal circuit. We show that microglia regulate pruning of retinal ganglion cell axons and are important for proper behavioral response to dark and bright looming stimuli. We identify amphibian regulator of complement activation 3, a homolog of human CD46, as a neuronally-expressed synapse-associated complement inhibitory molecule that inhibits trogocytosis and axonal pruning. Using a membrane-bound complement C3 fusion protein, we demonstrate that enhancing complement activity enhances axonal pruning. Our results support the model that microglia remodel axons via trogocytosis and that neurons can control this process through expression of complement inhibitory proteins.
- Published
- 2021
154. Interleukin-1β and cathepsin D modulate formation of the terminal complement complex in cultured human disc tissue
- Author
-
Uwe Max Mauer, Raquel Gonçalves, Cornelia Neidlinger-Wilke, Rolf E Brenner, Graciosa Q. Teixeira, Zhiyao Yong, Amelie Kuhn, Michael Ruf, Markus Huber-Lang, Jana Riegger, and Anita Ignatius
- Subjects
Adolescent ,medicine.medical_treatment ,Interleukin-1beta ,Cathepsin D ,Inflammation ,CD59 ,Complement Membrane Attack Complex ,Intervertebral Disc Degeneration ,03 medical and health sciences ,Tissue culture ,chemistry.chemical_compound ,0302 clinical medicine ,medicine ,Humans ,Orthopedics and Sports Medicine ,Intervertebral Disc ,Cells, Cultured ,030304 developmental biology ,0303 health sciences ,business.industry ,CD46 ,Zymosan ,Molecular biology ,Complement system ,Cytokine ,chemistry ,Surgery ,medicine.symptom ,business ,030215 immunology - Abstract
Purpose Formation of terminal complement complex (TCC), a downstream complement system activation product inducing inflammatory processes and cell lysis, has been identified in degenerated discs. However, it remains unclear which molecular factors regulate complement activation during disc degeneration (DD). This study investigated a possible involvement of the pro-inflammatory cytokine interleukin-1β (IL-1β) and the lysosomal protease cathepsin D (CTSD). Methods Disc biopsies were collected from patients suffering from DD (n = 43) and adolescent idiopathic scoliosis (AIS, n = 13). Standardized tissue punches and isolated cells from nucleus pulposus (NP), annulus fibrosus (AF) and endplate (EP) were stimulated with 5% human serum (HS) alone or in combination with IL-1β, CTSD or zymosan. TCC formation and modulation by the complement regulatory proteins CD46, CD55 and CD59 were analysed. Results In DD tissue cultures, IL-1β stimulation decreased the percentage of TCC + cells in AF and EP (P P P P Conclusion These results suggest a functional relevance of IL-1β and CTSD in modulating TCC formation in DD, with differences between tissue regions. Although strong TCC deposition may represent a degeneration-associated event, IL-1β may inhibit it. In contrast, TCC formation was shown to be triggered by CTSD, indicating a multifunctional involvement in disc pathophysiology.
- Published
- 2021
155. Complement Regulation in Human Tenocytes under the Influence of Anaphylatoxin C5a
- Author
-
Jingjian Shi, Benjamin Kohl, Sandeep Silawal, and Gundula Schulze-Tanzil
- Subjects
Male ,0301 basic medicine ,complement regulation ,Time Factors ,lcsh:Chemistry ,0302 clinical medicine ,Gene expression ,tendinopathy ,Complement Activation ,lcsh:QH301-705.5 ,Spectroscopy ,CD55 Antigens ,Chemistry ,Interleukin ,General Medicine ,Computer Science Applications ,Tumor necrosis factor alpha ,Adult ,medicine.medical_specialty ,C5a ,Complement C5a ,chemical and pharmacologic phenomena ,CD59 ,anaphylatoxin ,Article ,Catalysis ,Recombinant C5a ,Inorganic Chemistry ,03 medical and health sciences ,Internal medicine ,medicine ,Humans ,Anaphylatoxin ,Physical and Theoretical Chemistry ,Receptor, Anaphylatoxin C5a ,Molecular Biology ,complement system ,tenocytes ,Interleukin-6 ,Tumor Necrosis Factor-alpha ,CD46 ,Organic Chemistry ,Complement System Proteins ,030229 sport sciences ,Complement system ,030104 developmental biology ,Endocrinology ,Gene Expression Regulation ,lcsh:Biology (General) ,lcsh:QD1-999 - Abstract
A central part of the complement system, the anaphylatoxin C5a was investigated in this study to learn its effects on tenocytes in respect to understanding the potential expression of other crucial complement factors and pro-inflammatory mediators involved in tendinopathy. Human hamstring tendon-derived tenocytes were treated with recombinant C5a protein in concentrations of 25 ng/mL and 100 ng/mL for 0.5 h (early phase), 4 h (intermediate phase), and 24 h (late phase). Tenocytes survival was assessed after 24 h stimulation by live-dead assay. The gene expression of complement-related factors C5aR, the complement regulatory proteins (CRPs) CD46, CD55, CD59, and of the pro-inflammatory cytokines tumor necrosis factor (TNF)-α and interleukin (IL)-6 was monitored using qPCR. Tenocytes were immunolabeled for C5aR and CD55 proteins. TNFα production was monitored by ELISA. Tenocyte survival was not impaired through C5a stimulation. Interestingly, the gene expression of C5aR and that of the CRPs CD46 and CD59 was significantly reduced in the intermediate and late phase, and that of TNFα only in an early phase, compared to the control group. ELISA analysis indicated a concomitant not significant trend of impaired TNFα protein synthesis at 4 h. However, there was also an early significant induction of CD55 and CD59 mediated by 25 ng/mL anaphylatoxin C5a. Hence, exposure of tenocytes to C5a obviously evokes a time and concentration-dependent response in their expression of complement and pro-inflammatory factors. C5a, released in damaged tendons, might directly contribute to tenocyte activation and thereby be involved in tendon healing and tendinopathy.
- Published
- 2021
156. Treatment of Human Pancreatic Cancers Following Local and Systemic Administration of Oncolytic Adenovirus Serotype 35.
- Author
-
Ono R, Takayama K, Onishi R, Tokuoka S, Sakurai F, and Mizuguchi H
- Subjects
- Mice, Animals, Humans, Serogroup, Mice, Nude, Adenoviridae genetics, Antibodies, Neutralizing, Cell Line, Tumor, Genetic Vectors, Pancreatic Neoplasms therapy, Oncolytic Virotherapy, Oncolytic Viruses genetics
- Abstract
Background/aim: Oncolytic adenoviruses (Ads) (OAds) are gaining attention as an effective remedy for pancreatic cancer. Most OAds are based on human Ad serotype 5 (Ad5) (OAd5); however, two major drawbacks of OAd5 have been reported. Expression of coxsackievirus-adenovirus receptor, a primary infection receptor of Ad5, is often decreased on malignant tumor cells, including pancreatic cancers. More than 60% of adults have neutralizing antibodies against Ad5. Previously, we developed an OAd composed of Ad serotype 35 (Ad35) (OAd35). Ad35 recognizes CD46, which is often up-regulated on pancreatic cancers. In addition, only 20% or fewer adults have anti-Ad35 neutralizing antibodies., Materials and Methods: We examined the tumor cell lysis activities of OAd35 in the four human pancreatic cancer cell lines in the presence and absence of human serum. The tumor growth suppression effects of OAd35 after local and systemic administration were evaluated in nude mice bearing human pancreatic tumors., Results: OAd35 showed higher levels of tumor cell lysis activities than OAd5 in the human pancreatic cancer cell lines AsPC-1 and BxPC-3. Although the in vitro tumor cell lysis activities of OAd5 against MIA PaCa-2 and PANC-1 cells were strongly attenuated in the presence of human serum, OAd35 mediated comparable levels of tumor cell lysis in the presence and absence of human serum. Systemic administration of OAd5 did not mediate significant growth inhibition against the subcutaneous BxPC-3 tumor. On the other hand, OAd35 significantly suppressed tumor growth., Conclusion: OAd35 would be suitable as an alternative anticancer agent for pancreatic cancer., (Copyright © 2023 International Institute of Anticancer Research (Dr. George J. Delinasios), All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
157. Genome-wide association meta-analysis for early age-related macular degeneration highlights novel loci and insights for advanced disease
- Author
-
Winkler, Thomas W., Grassmann, Felix, Brandl, Caroline, Kiel, Christina, Günther, Felix, Strunz, Tobias, Weidner, Lorraine, Zimmermann, Martina E., Korb, Christina A., Poplawski, Alicia, Schuster, Alexander K., Müller-Nurasyid, Martina, Peters, Annette, Rauscher, Franziska G., Elze, Tobias, Horn, Katrin, Scholz, Markus, Cañadas-Garre, Marisa, McKnight, Amy Jayne, Quinn, Nicola, Hogg, Ruth E., Küchenhoff, Helmut, Heid, Iris M., Stark, Klaus J., and Weber, Bernhard H. F.
- Published
- 2020
- Full Text
- View/download PDF
158. Functional characterization of two novel non-synonymous alterations in CD46 and a Q950H change in factor H found in atypical hemolytic uremic syndrome patients.
- Author
-
Mohlin, Frida C., Nilsson, Sara C., Levart, Tanja Kersnik, Golubovic, Ema, Rusai, Krisztina, Müller-Sacherer, Thomas, Arbeiter, Klaus, Pállinger, Éva, Szarvas, Nóra, Csuka, Dorottya, Szilágyi, Ágnes, Villoutreix, Bruno O., Prohászka, Zoltán, and Blom, Anna M.
- Subjects
- *
HEMOLYTIC-uremic syndrome , *INPATIENT care , *ONTOLOGY , *SICK people , *MEDICAL care - Abstract
Atypical hemolytic uremic syndrome (aHUS) is a disease of complement dysregulation, characterized by hemolytic anemia, thrombocytopenia and acute renal failure. Mutations in complement inhibitors are major risk factors for development of aHUS. The three aHUS patients reported in this study had several previously identified alterations in complement inhibitors; e.g. risk haplotypes in CD46 and factor H but we also identified two novel heterozygous non-synonymous CD46 alterations (p.E142Q and p.G259V). Presence of G259V caused decreased expression of the recombinant mutant CD46 compared to wild type (WT). Western blot analysis showed that the majority of the expressed G259V protein was in the precursor form, suggesting that it is processed less efficiently than WT. Low CD46 expression on the surface of the patient's neutrophils confirmed the in vitro results. Further, G259V had a substantially impaired ability to act as a cofactor to factor I, in the degradation of both C3b and C4b. The E142Q mutant showed neither decreased expression nor impaired function. Two of the patients also had a heterozygous non-synonymous alteration in factor H (p.Q950H), reported previously in aHUS but not functionally tested. This variant showed moderately impaired function in hemolytic assays, both using patient sera and recombinant proteins. The recombinant Q950H also showed a somewhat decreased expression compared to WT but the complement inhibitory function in fluid phase was normal. Taken together, we report a novel CD46 alteration showing both a decreased protein expression and substantially impaired cofactor function (G259V) and another without an effect on expression or cofactor function (E142Q). Moreover, mild consequences of a previously reported aHUS associated rare variant in factor H (Q950H) was also revealed, underlining the clear need for functional characterization of each new aHUS associated mutation. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
159. Complement regulator CD46: genetic variants and disease associations.
- Author
-
Liszewski, M. Kathryn and Atkinson, John P.
- Abstract
Membrane cofactor protein (MCP; CD46) is an ubiquitously expressed complement regulatory protein that protects host cells from injury by complement. This type-I membrane glycoprotein serves as a cofactor for the serine protease factor I to mediate inactivation of C3b and C4b deposited on host cells. More than 60 disease-associated mutations in MCP have now been identified. The majority of the mutations are linked to a rare thrombotic microangiopathic-based disease, atypical hemolytic uremic syndrome (aHUS), but new putative links to systemic lupus erythematosus, glomerulonephritis, and pregnancy-related disorders among others have also been identified. This review summarizes our current knowledge of disease-associated mutations in this complement inhibitor. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
160. Adeno-associated virus mediated delivery of an engineered protein that combines the complement inhibitory properties of CD46, CD55 and CD59.
- Author
-
Leaderer, Derek, Cashman, Siobhan M., and Kumar‐Singh, Rajendra
- Abstract
Background A variety of disorders are associated with the activation of complement. CD46, CD55 and CD59 are the major membrane associated regulators of complement on human cells. Previously, we have found that independent expression of CD55, CD46 or CD59 through gene transfer protects murine tissues against human complement mediated attack. In the present study, we investigated the potential of combining the complement regulatory properties of CD46, CD55 and CD59 into single gene products expressed from an adeno-associated virus (AAV) vector in a soluble non-membrane anchored form. Methods Minigenes encoding the complement regulatory domains from CD46, CD55 and CD59 (SACT) or CD55 and CD59 (DTAC) were cloned into an AAV vector. The specific regulatory activity of each component of SACT and DTAC was measured in vitro. The recombinant AAV vectors were injected into the peritoneum of mice and the efficacy of the transgene products for being able to protect murine liver vasculature against human complement, specifically the membrane attack complex (MAC), was measured. Results SACT and DTAC exhibited properties similar to CD46, CD55 and CD59 or CD55 and CD59, respectively, in vitro. AAV mediated delivery of SACT or DTAC protected murine liver vasculature from human MAC deposition by 63.2% and 56.7%, respectively. Conclusions When delivered to mice in vivo via an AAV vector, SACT and DTAC are capable of limiting human complement mediated damage. SACT and DTAC merit further study as potential therapies for complement mediated disorders when delivered via a gene therapy approach. Copyright © 2015 John Wiley & Sons, Ltd. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
161. Recombinant Ad35 adenoviral proteins as potent modulators of human T-cell activation.
- Author
-
Hay, Joanne, Carter, Darrick, Lieber, André, and Astier, Anne L.
- Subjects
- *
ADENOVIRUSES , *T cells , *RECOMBINANT proteins , *IMMUNOLOGICAL adjuvants , *CD46 antigen , *COMPLEMENT (Immunology) - Abstract
The protein CD46 protects cells from complement attack by regulating cleavage of C3b and C3d. CD46 also regulates the adaptive immune response by controlling T-cell activation and differentiation. Co-engagement of the T-cell receptor and CD46 notably drives T-cell differentiation by switching production of interferon-γ to secretion of anti-inflammatory interleukin-10. This regulatory pathway is altered in several chronic inflammatory diseases, highlighting its key role for immune homeostasis. The manipulation of the CD46 pathway may therefore provide a powerful means to regulate immune responses. Herein, we investigated the effect of recombinant proteins derived from the fibre knob of the adenovirus serotype 35 (Ad35) that uses CD46 as its entry receptor, on human T-cell activation. We compared the effects of Ad35K++, engineered to exhibit enhanced affinity to CD46, and of Ad35K-, mutated in the binding site for CD46. Ad35K++ profoundly affects T-cell activation by decreasing the levels of CD46 at the surface of primary T cells, and impairing T-cell coactivation, shown by decreased CD25 expression, reduced proliferation and lower secretion of interleukin-10 and interferon-γ. In contrast, Ad35K- acts a potent co-activator of T cells, enhancing T-cell proliferation and cytokine production. These data show that recombinant Ad35 proteins are potent modulators of human T-cell activation, and support their further development as potential drugs targeting T-cell responses. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
162. Different impact of bovine complement regulatory protein 46 (CD46 bov ) as a cellular receptor for members of the species Pestivirus H and Pestivirus G .
- Author
-
Leveringhaus E, Cagatay GN, Hardt J, Becher P, and Postel A
- Subjects
- Animals, Bovine Virus Diarrhea-Mucosal Disease genetics, Bovine Virus Diarrhea-Mucosal Disease virology, Cattle, Cell Line, Diarrhea Viruses, Bovine Viral classification, Diarrhea Viruses, Bovine Viral genetics, Membrane Cofactor Protein genetics, Receptors, Virus genetics, Virus Internalization, Bovine Virus Diarrhea-Mucosal Disease metabolism, Diarrhea Viruses, Bovine Viral physiology, Membrane Cofactor Protein metabolism, Receptors, Virus metabolism
- Abstract
The genus Pestivirus within the family Flaviviridae comprises highly relevant animal pathogens such as bovine viral diarrhoea virus 1 and 2 (BVDV-1 and -2) classified into the two species Pestivirus A and Pestivirus B , respectively. First described in 2004, HoBi-like pestiviruses (HoBiPeV) represent emerging bovine pathogens that belong to a separate species ( Pestivirus H ), but share many similarities with BVDV-1 and -2. Additionally, two giraffe pestivirus (GPeV) strains both originating from Kenya represent another distinct species ( Pestivirus G ), whose members replicate very efficiently in bovine cells. In this study, we investigated the role of bovine complement regulatory protein 46 (CD46
bov ), the receptor of BVDV-1 and -2, in the entry of HoBiPeV and GPeV. For this purpose, bovine CD46-knockout and CD46-rescue cell lines were generated by CRISPR/Cas9 technology and subsequent trans-complementation, respectively. Our results provide strong evidence that the impact of CD46bov differs between viruses belonging to Pestivirus H and viruses representing Pestivirus G : CD46bov revealed to be a major cellular entry factor for HoBiPeV strain HaVi-20. In contrast, GPeV strain PG-2 presented as largely independent of CD46bov , suggesting a different entry mechanism involving other molecular determinants which remain to be identified. In addition, we demonstrated that, similar to BVDV-1 and -2, virus isolates of both Pestivirus H and Pestivirus G are able to adapt to cell culture conditions by using heparan sulfate to enter the host cell. In conclusion, our findings show that different bovine pestiviruses use diverse mechanisms of host cell entry.- Published
- 2022
- Full Text
- View/download PDF
163. Integrating Host Genomics with Surveillance for Invasive Bacterial Diseases
- Author
-
Dana C. Crawford, Shanta M. Zimmer, Craig A. Morin, Nancy E. Messonnier, Ruth Lynfield, Qian Yi, Cynthia Shephard, Michelle Wong, Mark J. Rieder, Robert J. Livingston, Deborah A. Nickerson, Cynthia G. Whitney, and Jairam Lingappa
- Subjects
Active bacterial core surveillance ,Neisseria meningitidis ,blood spots ,host gene ,CD46 ,dispatch ,Medicine ,Infectious and parasitic diseases ,RC109-216 - Abstract
We tested the feasibility of linking Active Bacterial Core surveillance, a prospective, population-based surveillance system for invasive bacterial disease, to a newborn dried blood spot (nDBS) repository. Using nDBS specimens, we resequenced CD46, putative host gene receptor for Neisseria meningitidis, and identified variants associated with susceptibility to this disease.
- Published
- 2008
- Full Text
- View/download PDF
164. Inherited Kidney Complement Diseases
- Author
-
Anne-Laure Lapeyraque, Damien Noone, Christoph Licht, Mathieu Lemaire, and Véronique Frémeaux-Bacchi
- Subjects
Epidemiology ,030232 urology & nephrology ,Complement factor I ,Critical Care and Intensive Care Medicine ,Bioinformatics ,Complement factor B ,03 medical and health sciences ,0302 clinical medicine ,Atypical hemolytic uremic syndrome ,medicine ,Humans ,030304 developmental biology ,Atypical Hemolytic Uremic Syndrome ,0303 health sciences ,Transplantation ,Complement component 3 ,CD46 ,business.industry ,Genomics of Kidney Disease ,Complement C3 ,medicine.disease ,Complement (complexity) ,Complement system ,Nephrology ,Factor H ,Complement Factor H ,Kidney Diseases ,business ,Complement Factor B - Abstract
In the past 20 years, we have witnessed tremendous advances in our ability to diagnose and treat genetic diseases of the kidney caused by complement dysregulation. Staggering progress was realized toward a better understanding of the genetic underpinnings and pathophysiology of many forms of atypical hemolytic uremic syndrome (aHUS) and C3-dominant glomerulopathies that are driven by complement system abnormalities. Many of these seminal discoveries paved the way for the design and characterization of several innovative therapies, some of which have already radically improved patients' outcomes. This review offers a broad overview of the exciting developments that have occurred in the recent past, with a particular focus on single-gene (or Mendelian), complement-driven aHUS and C3-dominant glomerulopathies that should be of interest to both nephrologists and kidney researchers. The discussion is restricted to genes with robust associations with both aHUS and C3-dominant glomerulopathies (complement factor H, complement component 3, complement factor H-related proteins) or only aHUS (complement factor B, complement factor I, and membrane cofactor protein). Key questions and challenges are highlighted, along with potential avenues for future directions.
- Published
- 2021
165. Treating Prostate Cancer by Antibody-Drug Conjugates
- Author
-
Rodolfo Montironi, Nadia Storti, Alessia Cimadamore, Matteo Santoni, Nicola Battelli, Veronica Mollica, Marina Scarpelli, Francesco Massari, Alessandro Rizzo, Matteo Rosellini, Rosellini M., Santoni M., Mollica V., Rizzo A., Cimadamore A., Scarpelli M., Storti N., Battelli N., Montironi R., and Massari F.
- Subjects
Male ,Oncology ,Immunoconjugates ,medicine.medical_treatment ,Review ,Disease ,lcsh:Chemistry ,Prostate cancer ,Antineoplastic Agents, Immunological ,Prostate ,Molecular Targeted Therapy ,CD46 ,lcsh:QH301-705.5 ,Spectroscopy ,Clinical Trials as Topic ,Disease Management ,Standard of Care ,General Medicine ,Prognosis ,prostate cancer ,Computer Science Applications ,Prostatic Neoplasms, Castration-Resistant ,Treatment Outcome ,medicine.anatomical_structure ,B7-H3 ,Disease Susceptibility ,medicine.medical_specialty ,Malignancy ,Catalysis ,Inorganic Chemistry ,Breast cancer ,Antibody drug conjugate ,Internal medicine ,Biomarkers, Tumor ,medicine ,PSMA ,Animals ,Humans ,Physical and Theoretical Chemistry ,Molecular Biology ,STEAP1 ,antibody drug conjugates ,business.industry ,target therapy ,Organic Chemistry ,Prostatic Neoplasms ,Cancer ,Immunotherapy ,Antibody drug conjugates ,Target therapy ,TROP2 ,medicine.disease ,lcsh:Biology (General) ,lcsh:QD1-999 ,Cancer cell ,business - Abstract
Prostate cancer is the most frequent malignancy in the worldwide male population; it is also one of the most common among all the leading cancer-related death causes. In the last two decades, the therapeutic scenario of metastatic castration-resistant prostate cancer has been enriched by the use of chemotherapy and androgen receptor signaling inhibitors (ARSI) and, more recently, by immunotherapy and poly(ADP–ribose) polymerase (PARP) inhibitors. At the same time, several trials have shown the survival benefits related to the administration of novel ARSIs among patients with non-castration-resistant metastatic disease along with nonmetastatic castration-resistant cancer too. Consequently, the therapeutic course of this malignancy has been radically expanded, ensuring survival benefits never seen before. Among the more recently emerging agents, the so-called “antibody–drug conjugates” (ADCs) are noteworthy because of their clinical practice changing outcomes obtained in the management of other malignancies (including breast cancer). The ADCs are novel compounds consisting of cytotoxic agents (also known as the payload) linked to specific antibodies able to recognize antigens expressed over cancer cells’ surfaces. As for prostate cancer, researchers are focusing on STEAP1, TROP2, PSMA, CD46 and B7-H3 as optimal antigens which may be targeted by ADCs. In this paper, we review the pivotal trials that have currently changed the therapeutic approach to prostate cancer, both in the nonmetastatic castration-resistant and metastatic settings. Therefore, we focus on recently published and ongoing trials designed to investigate the clinical activity of ADCs against prostate malignancy, characterizing these agents. Lastly, we briefly discuss some ADCs-related issues with corresponding strategies to overwhelm them, along with future perspectives for these promising novel compounds.
- Published
- 2021
166. The Fiber Knob Protein of Human Adenovirus Type 49 Mediates Highly Efficient and Promiscuous Infection of Cancer Cell Lines Using a Novel Cell Entry Mechanism
- Author
-
Carly M. Bliss, Pierre J. Rizkallah, James A. Davies, Emily A. Bates, Alan L. Parker, Elise Moses, Gareth Marlow, Rosie M. Mundy, David K. Cole, and Alexander T. Baker
- Subjects
Genetic Vectors ,Immunology ,Population ,Coxsackievirus ,Microbiology ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,Neoplasms ,Virology ,Humans ,anticancer therapy ,Vector (molecular biology) ,Receptor ,education ,Tropism ,030304 developmental biology ,Oncolytic Virotherapy ,0303 health sciences ,education.field_of_study ,biology ,CD46 ,Adenoviruses, Human ,virus diseases ,biochemical phenomena, metabolism, and nutrition ,Virus Internalization ,surface receptor ,biology.organism_classification ,eye diseases ,Virus-Cell Interactions ,3. Good health ,Oncolytic virus ,Cell biology ,adenoviruses ,oncolytic viruses ,030220 oncology & carcinogenesis ,Insect Science ,Receptors, Virus ,Capsid Proteins ,Cellular Tropism - Abstract
Adenoviruses are powerful tools experimentally and clinically. To maximize efficacy, the development of serotypes with low preexisting levels of immunity in the population is desirable., The human adenovirus (HAdV) phylogenetic tree is diverse, divided across seven species and comprising over 100 individual types. Species D HAdV are rarely isolated with low rates of preexisting immunity, making them appealing for therapeutic applications. Several species D vectors have been developed as vaccines against infectious diseases, where they induce robust immunity in preclinical models and early phase clinical trials. However, many aspects of the basic virology of species D HAdV, including their basic receptor usage and means of cell entry, remain understudied. Here, we investigated HAdV-D49, which previously has been studied for vaccine and vascular gene transfer applications. We generated a pseudotyped HAdV-C5 presenting the HAdV-D49 fiber knob protein (HAdV-C5/D49K). This pseudotyped vector was efficient at infecting cells devoid of all known HAdV receptors, indicating HAdV-D49 uses an unidentified cellular receptor. Conversely, a pseudotyped vector presenting the fiber knob protein of the closely related HAdV-D30 (HAdV-C5/D30K), differing in four amino acids from HAdV-D49, failed to demonstrate the same tropism. These four amino acid changes resulted in a change in isoelectric point of the knob protein, with HAdV-D49K possessing a basic apical region compared to a more acidic region in HAdV-D30K. Structurally and biologically we demonstrate that HAdV-D49 knob protein is unable to engage CD46, while potential interaction with coxsackievirus and adenovirus receptor (CAR) is extremely limited by extension of the DG loop. HAdV-C5/49K efficiently transduced cancer cell lines of pancreatic, breast, lung, esophageal, and ovarian origin, indicating it may have potential for oncolytic virotherapy applications, especially for difficult to transduce tumor types. IMPORTANCE Adenoviruses are powerful tools experimentally and clinically. To maximize efficacy, the development of serotypes with low preexisting levels of immunity in the population is desirable. Consequently, attention has focused on those derived from species D, which have proven robust vaccine platforms. This widespread usage is despite limited knowledge in their basic biology and cellular tropism. We investigated the tropism of HAdV-D49, demonstrating that it uses a novel cell entry mechanism that bypasses all known HAdV receptors. We demonstrate, biologically, that a pseudotyped HAdV-C5/D49K vector efficiently transduces a wide range of cell lines, including those presenting no known adenovirus receptor. Structural investigation suggests that this broad tropism is the result of a highly basic electrostatic surface potential, since a homologous pseudotyped vector with a more acidic surface potential, HAdV-C5/D30K, does not display a similar pantropism. Therefore, HAdV-C5/D49K may form a powerful vector for therapeutic applications capable of infecting difficult to transduce cells.
- Published
- 2021
167. Living Related Donor Kidney Transplantation in Atypical HUS: When Should It Be Considered?
- Author
-
Neetika Garg, Tripti Singh, Didier A. Mandelbrot, and Meghna P. Kurup
- Subjects
Thrombotic microangiopathy ,business.industry ,CD46 ,030232 urology & nephrology ,General Medicine ,Complement factor I ,030204 cardiovascular system & hematology ,urologic and male genital diseases ,medicine.disease ,Complement factor B ,Kidney Transplantation ,Transplantation ,03 medical and health sciences ,0302 clinical medicine ,hemic and lymphatic diseases ,Factor H ,Atypical hemolytic uremic syndrome ,Immunology ,Hemolytic-Uremic Syndrome ,Alternative complement pathway ,Living Donors ,Medicine ,Humans ,business ,Brief Communications - Abstract
Atypical hemolytic uremic syndrome (aHUS) is a thrombotic microangiopathy that manifests as hemolytic anemia, thrombocytopenia, and AKI. aHUS has been linked to the dysregulation of the alternative pathway of complement (1). In up to 60% of patients with aHUS, a genetic mutation known to cause dysregulation of complement system is found (2). Mutations in complement factor H ( CFH ), CD46 (membrane cofactor protein), complement factor I ( CFI ), thrombomodulin/CD141 ( THBD ), complement factor B ( CFB ), and C3 have been previously reported in patients with aHUS (3). People with mutations may not develop aHUS until adulthood (4). This incomplete penetrance indicates that a genetic mutation alone is insufficient to cause aHUS; instead, carriers of complement mutations require an additional trigger, such as pregnancy, viral infection, cancer, surgery, or medications, to progress to clinical disease of aHUS (5). As the type of mutation can influence both phenotype and rates of recurrence, genetic screening is advised pretransplant in patients with aHUS-related ESKD (2). However, there is no clear guidance for the genetic screening of family members who are potential living related donors. Historically, living related transplantation is actively discouraged in cases of aHUS due to increased risk of disease recurrence in recipients and risk of de novo disease in the donors (3,6). The recent advancement in genetics of aHUS challenges the recommendation against living related kidney transplantation. We present three patients with aHUS to explore whether living related kidney transplant can be considered as an option. …
- Published
- 2021
168. Primary Immunodeficiencies of Complement
- Author
-
Peter D. Arkwright
- Subjects
CD46 ,business.industry ,Complement factor I ,Eculizumab ,medicine.disease ,Complement factor B ,Membranoproliferative glomerulonephritis ,Immunology ,medicine ,Properdin ,business ,Immune complex disease ,Immunodeficiency ,medicine.drug - Abstract
Primary complement deficiencies are a complex and diverse group of over 20 distinct inherited immunodeficiency disorders. They make up 2–5% of all primary immunodeficiencies. These diseases can be divided into three broad groups. Classical pathway defects (C1, C2, C4) are associated with an increased risk of invasive pneumococcal infection and, in the case of C1q deficiency, an 80% risk of SLE-like immune complex disease. C1q deficiency has a 20% mortality rate in the first 20 years of life. Terminal complement defects (C5–C9) are largely associated with an increased risk of invasive meningococcal disease. Infection risk can be partly mitigated by vaccination. A third group includes deficiency (C3 loss of function, Factor B, properdin) or dysregulation of the C3 activity (C3 gain of function, Factor H, Factor I, membrane cofactor protein), the latter leads to endothelial damage and renal disease (aHUS or membranoproliferative glomerulonephritis) with a high risk of renal failure requiring dialysis. For most patients with primary complement deficiencies, CH50 and AH50 rather than C3 and C4, should be requested as the initial screening test. Management varies, ranging from augmenting pneumococcal and meningococcal antibody responses by immunization, antibiotic prophylaxis, immunosuppressants and HSCT for C1q deficiency, and anti-C5 monoclonal therapy for patients with aHUS.
- Published
- 2021
169. Differential Gene Expression in Bladder Tumors from Workers Occupationally Exposed to Arylamines
- Author
-
Zongli Xu, Jack A. Taylor, Ramya T Kolli, and Vijayalakshmi Panduri
- Subjects
Adult ,Male ,Article Subject ,DNA damage ,Gene Expression ,General Biochemistry, Genetics and Molecular Biology ,Risk Factors ,2-Naphthylamine ,Occupational Exposure ,Gene expression ,Humans ,Medicine ,Amines ,Gene ,Carcinogen ,Bladder cancer ,General Immunology and Microbiology ,business.industry ,CD46 ,Benzidines ,Cancer ,General Medicine ,Middle Aged ,medicine.disease ,Urinary Bladder Neoplasms ,Case-Control Studies ,YWHAZ ,Carcinogens ,Cancer research ,business ,Research Article - Abstract
Occupational exposure to the arylamines benzidine and β-naphthylamine increase bladder cancer risk up to 100-fold, making them some of the most powerful human carcinogens. We hypothesize that tumors arising in people with occupational exposures have different patterns of gene expression than histologically similar tumors from people without such exposures. In a case-case study, we compare gene expression in 22 formalin-fixed paraffin-embedded (FFPE) bladder tumors from men with high-level occupational exposure to arylamines to that in 26 FFPE bladder tumors from men without such exposure. Gene expression analysis was performed on the NanoString nCounter system using a PanCancer Progression Panel comprised of 740 cancer progression-related genes and a custom panel of 69 arylamine- and bladder cancer-related genes which were chosen from in vitro studies. Although fold differences were small, there was evidence of differential expression by exposure status for 17 genes from the Progression Panel and 4 genes from the custom panel. In total, 10 genes showed dose-response association at a p < 0.01 , of which 4 genes (CD46, NR4A1, BAX, and YWHAZ) passed a false discovery rate (FDR) q value cutoff of 0.05 but were not significant after Bonferroni correction. Overall, we find limited evidence for differentially expressed genes in pathways related to DNA damage signaling and epithelial-to-mesenchymal transition (EMT).
- Published
- 2021
- Full Text
- View/download PDF
170. COVID-19 Vaccines and Thrombosis-Roadblock or Dead-End Street?
- Author
-
Elrashdy M. Redwan, Ramesh Kandimalla, Damiano Pizzol, Murtaza M. Tambuwala, Debmalya Barh, Yogendra Kumar Mishra, Vladimir N. Uversky, Wagner Baetas-da-Cruz, Bruce D. Uhal, Pritam Kumar Panda, Kenneth Lundstrom, Alaa A. A. Aljabali, Gaurav Chauhan, Samendra P. Sherchan, Amos Lal, Tarek Mohamed Abd El-Aziz, Nima Rezaei, Nicolas G. Bazan, Pabitra Pal Choudhury, Adam Brufsky, Sk. Sarif Hassan, Parise Adadi, Giorgio Palù, Ángel Serrano-Aroca, Gajendra Kumar Azad, and Kazuo Takayama
- Subjects
Opinion ,Infectious Medicine ,COVID-19 Vaccines ,Infektionsmedicin ,030204 cardiovascular system & hematology ,medicine.disease_cause ,Biochemistry ,Microbiology ,Viral vector ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Risk Factors ,ChAdOx1 nCoV-19 ,Humans ,Medicine ,030212 general & internal medicine ,Adverse effect ,Molecular Biology ,BNT162 Vaccine ,thrombosis ,Coronavirus ,Smokers ,Ad26COVS1 ,biology ,CD46 ,business.industry ,SARS-CoV-2 ,Vaccination ,COVID-19 ,vaccines ,Thrombocytopenia ,QR1-502 ,chronic smokers ,Spike Glycoprotein, Coronavirus ,Immunology ,biology.protein ,Antibody ,business ,Platelet factor 4 ,2019-nCoV Vaccine mRNA-1273 - Abstract
Two adenovirus-based vaccines, ChAdOx1 nCoV-19 and Ad26.COV2.S, and two mRNA-based vaccines, BNT162b2 and mRNA.1273, have been approved by the European Medicines Agency (EMA), and are invaluable in preventing and reducing the incidence of coronavirus disease-2019 (COVID-19). Recent reports have pointed to thrombosis with associated thrombocytopenia as an adverse effect occurring at a low frequency in some individuals after vaccination. The causes of such events may be related to SARS-CoV-2 spike protein interactions with different C-type lectin receptors, heparan sulfate proteoglycans (HSPGs) and the CD147 receptor, or to different soluble splice variants of the spike protein, adenovirus vector interactions with the CD46 receptor or platelet factor 4 antibodies. Similar findings have been reported for several viral diseases after vaccine administration. In addition, immunological mechanisms elicited by viral vectors related to cellular delivery could play a relevant role in individuals with certain genetic backgrounds. Although rare, the potential COVID-19 vaccine-induced immune thrombotic thrombocytopenia (VITT) requires immediate validation, especially in risk groups, such as the elderly, chronic smokers, and individuals with pre-existing incidences of thrombocytopenia; and if necessary, a reformulation of existing vaccines.
- Published
- 2021
171. Xenotransplantation 1.0 to 2.0
- Author
-
Christiane Ferran, Omar J. Haque, Daniel J Cloonan, and Erin E. McIntosh
- Subjects
Transplantation ,Gene knockdown ,Immune system ,Genome editing ,CD46 ,business.industry ,Xenotransplantation ,medicine.medical_treatment ,Immunology ,medicine ,Endogenous retrovirus ,business ,Ex vivo - Abstract
Xenotransplantation, mostly from pig donors, represents a major untapped source to mitigate the shortage of organs suitable for transplantation. This chapter outlines the major historic events that shaped the field of xenotransplantation, from the advent of cyclosporine A, to genetic manipulations to overcome hyperacute rejection (HAR) through knockdown of α1,3-Galactosyl transferase and overexpression of human complement regulatory proteins (CRP), challenges posed by the discovery of porcine endogenous retroviruses, and the recent breakthrough of CRISPR-Cas9 gene editing. Beyond HAR, this chapter reviews the remaining immunologic barriers to successful xenotransplantation from acute vascular rejection/delayed xenograft rejection (AVR/DXR), to T-cell mediated acute rejection and chronic rejection. A special emphasis is placed on the pivotal role that activation of the xenograft endothelium plays in fueling AVR/DXR and its associated thrombotic microangiopathy, and on the identity of a handful of genes—chief amongst them A20—that can mitigate the pro-inflammatory and pro-coagulant crisis by allowing the endothelium to “accommodate” to the host immune and inflammatory attack. Revolutionary technologies that are enabling progress such as targeted gene disruption and transgenesis, CRISPR-Cas9 gene editing, cloning, and ex vivo machine perfusion systems are described. The chapter also proposes a wish list for engineering the best possible pig for xenotransplantation. A minimalistic, yet optimal, recipe supported by both in vitro and in vivo evidence calls for a pig that will be a triple knockout for the enzymes synthesizing xenogeneic carbohydrates, knockout for class I swine leukocyte antigen, and overexpressing human CRPs (CD46, CD55, or CD59) as well as regulators of coagulation (thrombomodulin, TFPI CD39), A20, HO-1, and immunoregulatory molecules such as human CD47. Finally, persistent challenges in xenotransplantation are acknowledged, including molecular incompatibilities between pigs and humans affecting immune regulation, coagulation and even organ function or animal viability, as well as ethical considerations and cost of engineering these organs.
- Published
- 2020
172. The Combination of gQ1 and gQ2 in Human Herpesvirus 6A and 6B Regulates the Viral Tetramer Function for Their Receptor Recognition
- Author
-
Mitsuhiro Nishimura, Lidya Handayani Tjan, Jun Arii, Anna Lystia Poetranto, Chisato Yamamoto, Aika Wakata, Akiko Kawabata, and Yasuko Mori
- Subjects
viruses ,Immunology ,Mutant ,Biology ,Microbiology ,Virus ,Tetramer ,Viral entry ,CD134 ,Virology ,gQ2 ,gQ1 ,Receptor ,CD46 ,chemistry.chemical_classification ,Cell fusion ,tetramer ,virus diseases ,biochemical phenomena, metabolism, and nutrition ,Cell biology ,Virus-Cell Interactions ,chemistry ,Insect Science ,HHV-6A ,Glycoprotein ,HHV-6B - Abstract
Human herpesvirus 6A (HHV-6A) and HHV-6B use different cellular receptors, human CD46 and CD134, respectively, and have different cell tropisms, although they have 90% similarity at the nucleotide level. An important feature that characterizes HHV-6A/6B is the glycoprotein H (gH)/gL/gQ1/gQ2 complex (a tetramer) that each virus has specifically on its envelope. Here, to determine which molecules in the tetramer contribute to the specificity for each receptor, we developed a cell-cell fusion assay system for HHV-6A and HHV-6B that uses cells expressing CD46 or CD134. With this system, when we replaced the gQ1 or gQ2 of HHV-6A with that of HHV-6B in the tetramer, the cell fusion activity mediated by glycoproteins via CD46 was lower than that seen with the original-type tetramer. When we replaced the gQ1 or the gQ2 of HHV-6A with that of HHV-6B in the tetramer, the cell fusion mediated by glycoproteins via CD134 was not seen. In addition, we generated two types of C-terminal truncation mutants of HHV-6A gQ2 (AgQ2) to examine the interaction domains of HHV-6A gQ1 (AgQ1) and AgQ2. We found that amino acid residues 163 to 185 in AgQ2 are important for interaction of AgQ1 and AgQ2. Finally, to investigate whether HHV-6B gQ2 (BgQ2) can complement AgQ2, an HHV-6A genome harboring BgQ2 was constructed. The mutant could not produce an infectious virus, indicating that BgQ2 cannot work for the propagation of HHV-6A. These results suggest that gQ2 supports the tetramer's function, and the combination of gQ1 and gQ2 is critical for virus propagation. IMPORTANCE Glycoprotein Q2 (gQ2), an essential gene for virus propagation, forms a heterodimer with gQ1. The gQ1/gQ2 complex has a critical role in receptor recognition in the gH/gL/gQ1/gQ2 complex (a tetramer). We investigated whether gQ2 regulates the specific interaction between the HHV-6A or -6B tetramer and CD46 or CD134. We established a cell-cell fusion assay system for HHV-6A/6B and switched the gQ1 or gQ2 of HHV-6A with that of HHV-6B in the tetramer. Although cell fusion was induced via CD46 when gQ1 or gQ2 was switched between HHV-6A and -6B, the activity was lower than that of the original combination. When gQ1 or gQ2 was switched in HHV-6A and -6B, no cell fusion was observed via CD134. HHV-6B gQ2 could not complement the function of HHV-6A's gQ2 in HHV-6A propagation, suggesting that the combination of gQ1 and gQ2 is critical for regulating the specificity of the tetramer's function for virus entry.
- Published
- 2020
173. Human species D adenovirus hexon capsid protein mediates cell entry through a direct interaction with CD46
- Author
-
Katja Mindler, Lijo John, Lars-Anders Carlson, B. David Persson, Michael Strebl, Menzo J. E. Havenga, Niklas Arnberg, Angelique A. C. Lemckert, Mónika Z. Ballmann, Lars Frängsmyr, Thilo Stehle, and Karim Rafie
- Subjects
Gene Expression Regulation, Viral ,Infectious Medicine ,COVID-19 Vaccines ,viruses ,receptor ,Cell ,hexon ,Infektionsmedicin ,Microbiology ,Cell Line ,Pharmaceutical Sciences ,vaccine ,medicine ,Humans ,Vector (molecular biology) ,Receptor ,Hexon protein ,CD46 ,Multidisciplinary ,Chemistry ,SARS-CoV-2 ,Adenoviruses, Human ,virus diseases ,adenovirus ,Biological Sciences ,Virus Internalization ,Farmaceutiska vetenskaper ,female genital diseases and pregnancy complications ,eye diseases ,Cell biology ,medicine.anatomical_structure ,Capsid ,Cell culture ,Capsid Proteins ,Function (biology) - Abstract
Significance The adenovirus capsid protein is built by three main capsomers: hexon, fiber, and penton base. Entry is mediated by fiber proteins anchoring the virus to host cell receptors and is followed by penton base proteins engaging coreceptors, resulting in entry. Here, we demonstrate that human adenovirus species D types, which constitute two-thirds of all human adenoviruses, enter host cells through a direct interaction between the hexon protein and CD46. This study provides insights into the entry mechanisms used by human adenoviruses. As these viruses are also used as vaccine vectors for prevention of other infectious diseases, the results provided will also be useful for further development of human adenovirus species D types as vaccine vectors., Human adenovirus species D (HAdV-D) types are currently being explored as vaccine vectors for coronavirus disease 2019 (COVID-19) and other severe infectious diseases. The efficacy of such vector-based vaccines depends on functional interactions with receptors on host cells. Adenoviruses of different species are assumed to enter host cells mainly by interactions between the knob domain of the protruding fiber capsid protein and cellular receptors. Using a cell-based receptor-screening assay, we identified CD46 as a receptor for HAdV-D56. The function of CD46 was validated in infection experiments using cells lacking and overexpressing CD46, and by competition infection experiments using soluble CD46. Remarkably, unlike HAdV-B types that engage CD46 through interactions with the knob domain of the fiber protein, HAdV-D types infect host cells through a direct interaction between CD46 and the hexon protein. Soluble hexon proteins (but not fiber knob) inhibited HAdV-D56 infection, and surface plasmon analyses demonstrated that CD46 binds to HAdV-D hexon (but not fiber knob) proteins. Cryoelectron microscopy analysis of the HAdV-D56 virion–CD46 complex confirmed the interaction and showed that CD46 binds to the central cavity of hexon trimers. Finally, soluble CD46 inhibited infection by 16 out of 17 investigated HAdV-D types, suggesting that CD46 is an important receptor for a large group of adenoviruses. In conclusion, this study identifies a noncanonical entry mechanism used by human adenoviruses, which adds to the knowledge of adenovirus biology and can also be useful for development of adenovirus-based vaccine vectors.
- Published
- 2020
174. Animal models of human herpesvirus 6 infection
- Author
-
Joséphine eReynaud and Branka eHorvat
- Subjects
HIV ,monkey ,Mouse ,CD46 ,animal model ,Neuroinflammation ,Microbiology ,QR1-502 - Abstract
Human herpesvirus (HHV)-6A and HHV-6B are two enveloped DNA viruses of β-herpesvirus family, infecting over 90% of the population and associated with several diseases, including exanthema subitum (for HHV-6B), multiple sclerosis and encephalitis, particularly in immunosuppressed patients. Animal models are highly important to better understand the pathogenesis of viral infections. Naturally developed neutralizing antibodies to HHV-6 or a related virus were found in different species of monkeys, suggesting their susceptibility to HHV-6 infection. Both HHV-6 DNA and infectious virus were detected in experimentally infected Cynomolgus and African green monkeys, although most animals remained clinically asymptomatic. Furthermore, HHV-6A infection was shown to accelerate the progression of AIDS in macaques and to lead to the development of neurological symptoms in the marmoset model. Humanized SCID mice efficiently replicated HHV-6 and were also susceptible to coinfection with HHV-6 and HIV-1. As CD46 was identified as a receptor for HHV-6, transgenic mice expressing human CD46 may present a potentially interesting model for study certain aspects of HHV-6 infection and neuroinflammation.
- Published
- 2013
- Full Text
- View/download PDF
175. The Host Cell Receptors for Measles Virus and Their Interaction with the Viral Hemagglutinin (H) Protein
- Author
-
Liang-Tzung Lin and Christopher D. Richardson
- Subjects
measles virus ,membrane cofactor protein ,CD46 ,signaling lymphocyte activation molecule family member 1 ,SLAMF1 ,SLAM ,CD150 ,nectin-4 ,polio virus receptor like protein 4 ,PVRL4 ,Microbiology ,QR1-502 - Abstract
The hemagglutinin (H) protein of measles virus (MeV) interacts with a cellular receptor which constitutes the initial stage of infection. Binding of H to this host cell receptor subsequently triggers the F protein to activate fusion between virus and host plasma membranes. The search for MeV receptors began with vaccine/laboratory virus strains and evolved to more relevant receptors used by wild-type MeV. Vaccine or laboratory strains of measles virus have been adapted to grow in common cell lines such as Vero and HeLa cells, and were found to use membrane cofactor protein (CD46) as a receptor. CD46 is a regulator that normally prevents cells from complement-mediated self-destruction, and is found on the surface of all human cells, with the exception of erythrocytes. Mutations in the H protein, which occur during adaptation and allow the virus to use CD46 as a receptor, have been identified. Wild-type isolates of measles virus cannot use the CD46 receptor. However, both vaccine/laboratory and wild-type strains can use an immune cell receptor called signaling lymphocyte activation molecule family member 1 (SLAMF1; also called CD150) and a recently discovered epithelial receptor known as Nectin-4. SLAMF1 is found on activated B, T, dendritic, and monocyte cells, and is the initial target for infections by measles virus. Nectin-4 is an adherens junction protein found at the basal surfaces of many polarized epithelial cells, including those of the airways. It is also over-expressed on the apical and basal surfaces of many adenocarcinomas, and is a cancer marker for metastasis and tumor survival. Nectin-4 is a secondary exit receptor which allows measles virus to replicate and amplify in the airways, where the virus is expelled from the body in aerosol droplets. The amino acid residues of H protein that are involved in binding to each of the receptors have been identified through X-ray crystallography and site-specific mutagenesis. Recombinant measles “blind” to each of these receptors have been constructed, allowing the virus to selectively infect receptor specific cell lines. Finally, the observations that SLAMF1 is found on lymphomas and that Nectin-4 is expressed on the cell surfaces of many adenocarcinomas highlight the potential of measles virus for oncolytic therapy. Although CD46 is also upregulated on many tumors, it is less useful as a target for cancer therapy, since normal human cells express this protein on their surfaces.
- Published
- 2016
- Full Text
- View/download PDF
176. Crosstalk between TGF-β1 and complement activation augments epithelial injury in pulmonary fibrosis.
- Author
-
Hongmei Gu, Mickler, Elizabeth A., Cummings, Oscar W., Sandusky, George E., Weber, Daniel J., Gracon, Adam, Woodruff, Trent, Wilkes, David S., and Vittal, Ragini
- Subjects
- *
IDIOPATHIC pulmonary fibrosis , *TRANSFORMING growth factors-beta , *PULMONARY fibrosis , *LUNG diseases , *IDIOPATHIC interstitial pneumonias - Abstract
The epithelial complement inhibitory proteins (CIPs) cluster of differentiation 46 and 55 (CD46 and CD55) regulate circulating immune complex-mediated complement activation in idiopathic pulmonary fibrosis (IPF). Our previous studies demonstrated that IL-17A mediates epithelial injury via transforming growth factor β1 (TGF-β1) and down-regulates CIPs. In the current study, we examined the mechanistic role of TGF-β1 in complement activation-mediated airway epithelial injury in IPF pathogenesis. We observed lower epithelial CIP expression in IPF lungs compared to normal lungs, associated with elevated levels of complement component 3a and 5a (C3a and C5a), locally and systemically. In normal primary human small airway epithelial cells (SAECs) treated with TGF-β1 (10 ng/ml), C3a, or C5a (100 nM), we observed loss of CIPs and increased poly(ADP-ribose) polymerase (PARP) activation [also observed with RNA interference (RNAi) of CD46/CD55]. TGF-β1-mediated loss of CIPs and Snail induction [SNAI1; a transcriptional repressor of E-cadherin (E-CAD)J was blocked by inhibiting mitogen-activated protein kinase (p38MAPK; SB203580) and RNAi silencing of SNAI1. C3a- and C5a-mediated loss of CIPs was also blocked by p38MAPK inhibition. While C3a upregulated TGFb transcripts, both C3a and C5a down-regulated SMAD7 (negative regulator of TGF-β), and whereas TGF-β1 induced C3a/C5a receptor (C3aR/C5aR) expression, pharmacologic C3aR/C5aR inhibition protected against C3a-/C5a-mediated loss of CIPs. Taken together, our results suggest that epithelial injury in IPF can be collectively amplified as a result of TGF-β1-induced loss of CIPs leading to complement activation that down-regulates CIPs and induces TGF-β1 expression. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
177. Spontaneous abortion is associated with elevated systemic C5a and reduced mRNA of complement inhibitory proteins in placenta.
- Author
-
Banadakoppa, M., Chauhan, M. S., Havemann, D., Balakrishnan, M., Dominic, J. S., and Yallampalli, C.
- Subjects
- *
MISCARRIAGE , *PLACENTA diseases , *PREGNANCY complications , *COMPLEMENT activation , *INFLAMMATION , *VASCULAR endothelial growth factors , *MILIEU therapy - Abstract
Spontaneous abortion in early pregnancy due to unknown reasons is a common problem. The excess complement activation and consequent placental inflammation and anti-angiogenic milieu is emerging as an important associated factor in many pregnancy-related complications. In the present study we sought to examine the expression of complement inhibitory proteins at the feto-maternal interface and levels of complement split products in the circulation to understand their role in spontaneous abortion. Consenting pregnant women who either underwent elective abortion due to non-clinical reasons ( n = 13) or suffered miscarriage ( n = 14) were recruited for the study. Systemic levels of complement factors C3a and C5a were measured by enzyme-linked immunosorbent assay ( ELISA). Plasma C5 and C3 protein levels were examined by Western blot. Expressions of complement regulatory proteins such as CD46 and CD55 in the decidua were investigated by quantitative polymerase chain reaction ( PCR) and Western blot. The median of plasma C3a level was 82·83 ng/ml and 66·17 ng/ml in elective and spontaneous abortion patients, respectively. Medians of plasma C5a levels in elective and spontaneous abortion patients were 0·96 ng/ml and 1·14 ng/ml, respectively. Only plasma C5a levels but not C3a levels showed significant elevation in spontaneous abortion patients compared to elective abortion patients. Further, there was a threefold decrease in the mRNA expressions of complement inhibitory proteins CD46 and CD55 in the decidua obtained from spontaneous abortion patients compared to that of elective abortion patients. These data suggested that dysregulated complement cascade may be associated with spontaneous abortion. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
178. The complement receptors CD46, CD55 and CD59 are regulated by the tumour microenvironment of head and neck cancer to facilitate escape of complement attack.
- Author
-
Kesselring, Rebecca, Thiel, Annette, Pries, Ralph, Fichtner-Feigl, Stefan, Brunner, Stefan, Seidel, Philipp, Bruchhage, Karl-Ludwig, and Wollenberg, Barbara
- Subjects
- *
LYMPHOCYTES , *CELL receptors , *ACADEMIC medical centers , *COMPLEMENT (Immunology) , *FLOW cytometry , *HEAD tumors , *NECK tumors , *SQUAMOUS cell carcinoma , *WESTERN immunoblotting , *PHYSIOLOGY , *CELL physiology - Abstract
Background Membrane-bound complement restriction proteins (mCRPs) CD46, CD55 and CD59 enable tumour cells to evade complement dependent cytotoxicity and antibody-dependent killing mechanisms. But less is known about the role of these mCRPs in head and neck cancer. Methods In this study we determined the expression of the mCRPs on head and neck squamous cell carcinoma (HNSCC) cell lines, on tumour tissue and TDLNs (tumour-draining lymph nodes) as well as on lymphocytes from HNSCC patients. The influence of the HNSCC microenvironment on the mCRP regulation was analysed using Flow Cytometry, Western blotting and small interfering RNAs (siRNA) transfection studies. Results We examined the effects of the HNSCC tumour milieu on the expression levels of CD46, CD55 and CD59. We investigated the susceptibility of HNSCC cells to CDC (complement-dependent cytotoxicity) while silencing the mCRPs. Our results demonstrate a huge influence of the HNSCC tumour microenvironment on the regulation of mCRP expression and show a reciprocal regulation between the different mCRPs themselves. Conclusions In summary, our data indicate that HNSCC has evolved different strategies to evade complement attacks and that the tumour microenvironment leads to the enhancement of complement resistance of the surrounding tissue. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
179. Uromodulin aggravates renal tubulointerstitial injury through activation of the complement pathway in rats
- Author
-
Maojing Liu, Xiangdong Yang, Qiaoling Sun, Fei Shen, Zhao Hu, Li Yu, and Fei Pei
- Subjects
0301 basic medicine ,Male ,Pathology ,medicine.medical_specialty ,Tamm–Horsfall protein ,Physiology ,Clinical Biochemistry ,urologic and male genital diseases ,Cell Line ,Rats, Sprague-Dawley ,03 medical and health sciences ,Complement inhibitor ,0302 clinical medicine ,Western blot ,Fibrosis ,Uromodulin ,medicine ,Renal fibrosis ,Animals ,Humans ,Complement Activation ,Kidney ,biology ,medicine.diagnostic_test ,business.industry ,CD46 ,Cell Biology ,Complement System Proteins ,medicine.disease ,Complement system ,Rats ,030104 developmental biology ,medicine.anatomical_structure ,Kidney Tubules ,030220 oncology & carcinogenesis ,biology.protein ,Nephritis, Interstitial ,business ,Ureteral Obstruction - Abstract
Uromodulin (Umod) is the most abundant constituent of urine in humans and exclusively found in the kidney tubular epithelium. However, the specific role of Umod in renal tubulointerstitial injury is yet to be understood. The present study was conducted with aim of investigating the potential therapeutic mechanism of Umod in the regulation of renal tubulointerstitial injury. Protein expression of Umod in renal tubular epithelial cells was measured with the conduction of Western blot analysis. Enzyme-linked immunosorbent assay and immunofluorescence assay were performed to detect the complement activation products and the activation products of surface deposition. The expression of C1q, C2, C4, B factor, C3, C5, H factor, CD46, CD55, C3aR, and C5aR were determined with the use of reverse-transcription quantitative polymerase chain reaction and Western blot analyses. Subsequently, the unilateral ureteral obstruction (UUO) rat model was established. Renal tubulointerstitial injury was assessed with the application of hematoxylin-eosin staining and Masson staining in rats. UUO rats and normal rats were injected with si-NC or si-Umod and complement inhibitor. UUO rats were observed to have serious impairment of kidney tubule, renal tubular dilation, and epithelial atrophy, with downregulated Umod and activated complement pathway. Silencing of Umod resulted in the activation of complement system while promoting interstitial fibrosis in renal tubules. Moreover, addition of complement inhibitor significantly alleviated the renal tubule injury and fibrosis. Collectively, our study suggests that silencing of Umod mediates the complement pathway, exacerbating renal tubulointerstitial injury in rats, which provides insight into the development of novel therapeutic agents for renal tubulointerstitial injury.
- Published
- 2020
180. A SNP in intron 8 of CD46 causes a novel transcript associated with mastitis in Holsteins.
- Author
-
Xiuge Wang, Jifeng Zhong, Yundong Gao, Zhihua Ju, and Jinming Huang
- Abstract
Background: The membrane protein CD46, a ubiquitous cell surface pathogen receptor, can bind Streptococcus to trigger cell autophagy, which is a critical step in the control of infection. Results: In this study, we found a new splice variant designated CD46 transcript variant (CD46-TV). The splice variant is characterized by the retention of a 48 bp sequence from intron 8 of the bovine CD46 gene, which encodes a putative protein enlarged by 16 amino acids. CD46-TV mRNA was found to be over expressed in mastitis-infected mammary gland tissues relative to healthy tissues. A single nucleotide polymorphism (c. 1033 + 2184 C > T) in the exonic splicing enhancer (ESE) motif region was shown to result in the CD46-TV aberrant splice variant through constructing alternative alleles using the pSPL3 exon capturing vector and transfecting these into 293 T cells. Allelic frequency in 56,682 individuals belonging to 112 Bos taurus, Bos indicus, Bos javanicus, Bos grunniens and Bos mutus, etc. suggests that the C allele (80.09%) is the ancestral allele. Association analysis found that the mean genomic estimated breeding values (gEBV) for milk somatic cell score and the occurrence of clinical mastitis, as well as the milk somatic cell score of Chinese Holsteins with the CT genotype was lower than those of individuals with either the CC or TT genotypes. The mean gEBV for udder health synthesis for the TT genotype was greater than those for the CC or CT genotypes. Conclusions: Our findings suggest that the CD46 gene likely plays a critical role in the risk of mastitis caused by Streptococcus in dairy cows via an alternative splicing mechanism caused by a functional mutation in intron 8. Our data also underline the importance of variation within ESEs in regulating transcript processing. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
181. CD46 and Oncologic Interactions: Friendly Fire against Cancer
- Author
-
M. Kathryn Liszewski, John P. Atkinson, and Michelle Elvington
- Subjects
lcsh:Immunologic diseases. Allergy ,viruses ,Immunology ,Regulator ,antibody-drug conjugates ,Review ,Biology ,Malignant transformation ,Measles virus ,cancer therapeutics ,Drug Discovery ,medicine ,Immunology and Allergy ,complement ,CD46 ,membrane cofactor protein (MCP) ,Cancer ,adenovirus ,medicine.disease ,biology.organism_classification ,female genital diseases and pregnancy complications ,Complement system ,Oncolytic virus ,measles virus ,Monoclonal ,Cancer research ,lcsh:RC581-607 - Abstract
One of the most challenging aspects of cancer therapeutics is target selection. Recently, CD46 (membrane cofactor protein; MCP) has emerged as a key player in both malignant transformation as well as in cancer treatments. Normally a regulator of complement activation, CD46 is co-expressed as four predominant isoforms on almost all cell types. CD46 is highly overexpressed on a variety of human tumor cells. Clinical and experimental data support an association between increased CD46 expression and malignant transformation and metastasizing potential. Further, CD46 is a newly discovered driver of metabolic processes and plays a role in the intracellular complement system (complosome). CD46 is also known as a pathogen magnet due to its role as a receptor for numerous microbes, including several species of measles virus and adenoviruses. Strains of these two viruses have been exploited as vectors for the therapeutic development of oncolytic agents targeting CD46. In addition, monoclonal antibody-drug conjugates against CD46 also are being clinically evaluated. As a result, there are multiple early-phase clinical trials targeting CD46 to treat a variety of cancers. Here, we review CD46 relative to these oncologic connections.
- Published
- 2020
182. Molecular Studies and an ex vivo Complement Assay on Endothelium Highlight the Genetic Complexity of Atypical Hemolytic Uremic Syndrome: The Case of a Pedigree With a Null CD46 Variant
- Author
-
Marta Alberti, Paola Cuccarolo, Elisabetta Valoti, Caterina Mele, Paraskevas Iatropoulos, Miriam Galbusera, Ariela Benigni, Sara Gastoldi, Marina Noris, Rossella Piras, Marta Todeschini, Giuseppe Remuzzi, and Elena Bresin
- Subjects
0301 basic medicine ,Proband ,030232 urology & nephrology ,urologic and male genital diseases ,03 medical and health sciences ,splicing ,0302 clinical medicine ,hemic and lymphatic diseases ,Atypical hemolytic uremic syndrome ,Medicine ,complement ,Original Research ,Complement component 5 ,lcsh:R5-920 ,CD46 expression ,biology ,incomplete penetrance ,business.industry ,CD46 ,atypical hemolytic uremic syndrome ,Haplotype ,rare variants ,General Medicine ,medicine.disease ,Penetrance ,ex-vivo assay ,030104 developmental biology ,Immunology ,biology.protein ,Alternative complement pathway ,Antibody ,lcsh:Medicine (General) ,business ,membrane cofactor protein - Abstract
Atypical hemolytic uremic syndrome (aHUS) is an ultra-rare disease characterized by microangiopathic hemolysis, thrombocytopenia, and renal impairment and is associated with dysregulation of the alternative complement pathway on the microvascular endothelium. Outcomes have improved greatly with pharmacologic complement C5 blockade. Abnormalities in complement genes (CFH, CD46, CFI, CFB, C3, and THBD), CFH–CFHR genomic rearrangements, and anti-FH antibodies have been reported in 40–60% of cases. The penetrance of aHUS is incomplete in carriers of complement gene abnormalities; and multiple hits, including the CFH–H3 and CD46GGAAC risk haplotypes and the CFHR1*B risk allele, as well as environmental factors, contribute to disease development. Here, we investigated the determinants of penetrance of aHUS associated with CD46 genetic abnormalities. We studied 485 aHUS patients and found CD46 rare variants (RVs) in about 10%. The c.286+2T>G RV was the most prevalent (13/485) and was associated with G RV who experienced a severe form of aHUS and developed end-stage renal failure. The father and paternal uncle with the same variant in homozygosity and six heterozygous relatives are unaffected. Flow cytometry analysis showed about 50% reduction of CD46 expression on blood mononuclear cells from the heterozygous proband and over 90% reduction in cells from the proband's unaffected homozygous father and aunt. Further genetic studies did not reveal RVs in known aHUS-associated genes or common genetic modifiers that segregated with the disease. Importantly, a specific ex vivo test showed excessive complement deposition on endothelial cells exposed to sera from the proband, and also from his mother and maternal uncle, who do not carry the c.286+2T>G RV, indicating that they share a circulating defect that results in complement dysregulation on the endothelium. These results highlight the complexity of the genetics of aHUS and indicate that CD46 deficiency may not be enough to induce aHUS. We hypothesize that the proband inherited from his mother a genetic abnormality in a complement circulating factor that has not been identified yet, which synergized with the CD46 RV in predisposing him to the aHUS phenotype.
- Published
- 2020
183. Atypical HUS relapse triggered by Covid-19
- Author
-
Sabine Le Bot, Fadi Fakhouri, Véronique Frémeaux-Bacchi, Simon Ville, Agnès Chapelet-Debout, Clément Deltombe, and Gilles Blancho
- Subjects
2019-20 coronavirus outbreak ,Coronavirus disease 2019 (COVID-19) ,CD46 ,business.industry ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,SARS-CoV-2 infection ,atypical HUS ,medicine.disease ,Virology ,MCP mutation ,Complement system ,Nephrology ,Atypical hemolytic uremic syndrome ,Mutation (genetic algorithm) ,Severity of illness ,medicine ,business ,Letters to the Editor ,Covid-19 - Published
- 2020
184. Protein engineering to target complement evasion in cancer.
- Author
-
Carter, Darrick and Lieber, André
- Subjects
- *
PROTEIN engineering , *COMPLEMENT inhibition , *PROTEIN solubility , *CANCER cells , *CANCER invasiveness , *IMMUNOGLOBULINS - Abstract
Abstract: The complement system is composed of soluble factors in plasma that enhance or “complement” immune-mediated killing through innate and adaptive mechanisms. Activation of complement causes recruitment of immune cells; opsonization of coated cells; and direct killing of affected cells through a membrane attack complex (MAC). Tumor cells up-regulate complement inhibitory factors – one of several strategies to evade the immune system. In many cases as the tumor progresses, dramatic increases in complement inhibitory factors are found on these cells. This review focuses on the classic complement pathway and the role of major complement inhibitory factors in cancer immune evasion as well as on how current protein engineering efforts are being employed to increase complement fixing or to reverse complement resistance leading to better therapeutic outcomes in oncology. Strategies discussed include engineering of antibodies to enhance complement fixation, antibodies that neutralize complement inhibitory proteins as well as engineered constructs that specifically target inhibition of the complement system. [Copyright &y& Elsevier]
- Published
- 2014
- Full Text
- View/download PDF
185. Adenoviruses Types, Cell Receptors and Local Innate Cytokines in Adenovirus Infection.
- Author
-
Chen, Rong-Fu and Lee, Chun-Yi
- Subjects
- *
ADENOVIRUSES , *CYTOKINES , *PATHOGENIC microorganisms , *COXSACKIEVIRUSES , *IMMUNE response - Abstract
Adenovirus is a common infectious pathogen in both children and adults. It is a significant cause of morbidity in immunocompetent people living in crowded living conditions and of mortality in immunocompromised hosts. It has more recently become a popular vehicle for gene therapy applications. The host response to wild-type infection and gene therapy vector exposure involves both virus entry receptor and the innate immune systems. Cell-mediated recognition of viruses via capsid components has received significant attention, principally thought to be regulated by the coxsackievirus-adenovirus receptor (CAR), CD46, integrins and heparin sulfate-containing proteoglycans. Antiviral innate immune responses are initiated by the infected cell, which activates the interferon response to block viral replication, while simultaneously releasing chemokines to attract neutrophils and NK cells. This review discusses the innate immune response primarily during wild-type adenovirus infection because this serves as the basis for understanding the response during both natural infection and exposure to adenovirus vectors. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
186. Role of anti-CD40 antibody-mediated costimulation blockade on non-Gal antibody production and heterotopic cardiac xenograft survival in a GTKO.hCD46Tg pig-to-baboon model.
- Author
-
Mohiuddin, Muhammad M., Singh, Avneesh K., Corcoran, Philip C., Hoyt, Robert F., Thomas, Marvin L., Lewis, Billeta G. T., Eckhaus, Michael, Dabkowski, Nicole L., Belli, Aaron J., Reimann, Keith A., Ayares, David, and Horvath, Keith A.
- Subjects
- *
IMMUNOSUPPRESSION , *IMMUNOREGULATION , *IMMUNOLOGICAL tolerance , *IMMUNOGLOBULINS , *XENOGRAFTS , *XENOTRANSPLANTATION - Abstract
Background Recently, we have shown that an immunosuppression regimen including costimulation blockade via anti-CD154 antibody significantly prolongs the cardiac xenograft survival in a GTKO.hCD46Tg pig-to-baboon heterotopic xenotransplantation model. Unfortunately, many coagulation disorders were observed with the use of anti-CD154 antibody, and recipient survival was markedly reduced by these complications. Material and Methods In this experiment, we replaced anti-CD154 antibody with a more clinically acceptable anti-CD40 antibody while keeping the rest of the immunosuppressive regimen and the donor pig genetics the same. This was carried out to evaluate the antibody's role in xenograft survival and prevention of coagulopathies. Two available clones of anti-CD40 antibody were tested. One mouse anti-human CD40 antibody, (clone 3A8), activated B lymphocytes in vitro and only modestly suppressed antibody production in vivo. Whereas a recombinant mouse non-human primate chimeric raised against macaque CD40, (clone 2C10R4), blocked B-cell activation in vitro and completely blocked antibody production in vivo. Results The thrombotic complications seen with anti-CD154 antibody were effectively avoided but the graft survival, although extended, was not as prolonged as observed with anti-CD154 antibody treatment. The longest survival for the 3A8 antibody group was 27 days, and the longest graft survival in the 2C10R4 antibody group was 146 days. All of the grafts except two rejected and were explanted. Only two recipient baboons had to be euthanized due to unrelated complications, and the rest of the baboons remained healthy throughout the graft survival period or after graft explantation. In contrast to our anti-CD 154 antibody-treated baboons, the non-Gal antibody levels started to rise after B cells made their appearance around 8 weeks post-transplantation. Conclusions Anti-CD40 antibody at the current dose does not induce any coagulopathies but while effective, had reduced efficacy to induce similar long-term graft survival as with anti-CD154 antibody perhaps due to ineffective control of B-cell function and antibody production at the present dose. More experiments are required to determine antibody affinity and effective dose for inducing long-term cardiac xenograft survival. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
187. Adenovirus receptor expression in cancer and its multifaceted role in oncolytic adenovirus therapy
- Author
-
Lobke C M Hensen, Selas T F Bots, and Rob C. Hoeben
- Subjects
Oncolytic adenovirus ,Coxsackie and Adenovirus Receptor-Like Membrane Protein ,Combination therapy ,Receptor expression ,viruses ,Integrin ,oncolytic adenovirus therapy ,DSG-2 ,Review ,Catalysis ,Inorganic Chemistry ,lcsh:Chemistry ,Downregulation and upregulation ,Cell Line, Tumor ,Neoplasms ,receptor expression ,medicine ,Animals ,Humans ,Physical and Theoretical Chemistry ,human adenovirus ,Receptor ,Molecular Biology ,lcsh:QH301-705.5 ,CD46 ,Spectroscopy ,Oncolytic Virotherapy ,Desmoglein 2 ,biology ,business.industry ,Adenoviruses, Human ,Organic Chemistry ,Cancer ,General Medicine ,medicine.disease ,Combined Modality Therapy ,N-Acetylneuraminic Acid ,Computer Science Applications ,Oncolytic virus ,CAR ,Oncolytic Viruses ,lcsh:Biology (General) ,lcsh:QD1-999 ,sialic acid ,Cancer research ,biology.protein ,integrins ,Receptors, Virus ,business - Abstract
Oncolytic adenovirus therapy is believed to be a promising way to treat cancer patients. To be able to target tumor cells with an oncolytic adenovirus, expression of the adenovirus receptor on the tumor cell is essential. Different adenovirus types bind to different receptors on the cell, of which the expression can vary between tumor types. Pre-existing neutralizing immunity to human adenovirus species C type 5 (HAdV-C5) has hampered its therapeutic efficacy in clinical trials, hence several adenoviral vectors from different species are currently being developed as a means to evade pre-existing immunity. Therefore, knowledge on the expression of appropriate adenovirus receptors on tumor cells is important. This could aid in determining which tumor types would benefit most from treatment with a certain oncolytic adenovirus type. This review provides an overview of the known receptors for human adenoviruses and how their expression on tumor cells might be differentially regulated compared to healthy tissue, before and after standardized anticancer treatments. Mechanisms behind the up- or downregulation of adenovirus receptor expression are discussed, which could be used to find new targets for combination therapy to enhance the efficacy of oncolytic adenovirus therapy. Additionally, the utility of the adenovirus receptors in oncolytic virotherapy is examined, including their role in viral spread, which might even surpass their function as primary entry receptors. Finally, future directions are offered regarding the selection of adenovirus types to be used in oncolytic adenovirus therapy in the fight against cancer.
- Published
- 2020
188. Human Desmoglein-2 and Human CD46 Mediate Human Adenovirus Type 55 Infection, but Human Desmoglein-2 Plays the Major Roles
- Author
-
Chenchen Yang, Ying Feng, Xikui Sun, Linbing Qu, Jia Luo, Liqiang Feng, Xinglong Liu, Tao Shu, Changhua Yi, Xuehua Zheng, Guan Suhua, Mingli Hao, Wan Su, Ling Chen, X.L. Ye, Liang Li, and Liu Xiaolin
- Subjects
Adult ,Genetically modified mouse ,Immunology ,Mice, Transgenic ,CHO Cells ,Biology ,Microbiology ,Cell Line ,Membrane Cofactor Protein ,Pathogenesis ,Gene Knockout Techniques ,Mice ,Viral Proteins ,03 medical and health sciences ,Cricetulus ,RNA interference ,Virology ,Blocking antibody ,medicine ,Animals ,Humans ,Gene Knock-In Techniques ,Receptor ,Gene knockout ,030304 developmental biology ,Mice, Inbred BALB C ,0303 health sciences ,Desmoglein 2 ,030306 microbiology ,CD46 ,Adenoviruses, Human ,medicine.disease ,Virus-Cell Interactions ,Mice, Inbred C57BL ,Disease Models, Animal ,A549 Cells ,Gene Knockdown Techniques ,Insect Science ,Pneumonia (non-human) - Abstract
Human adenovirus type 55 (HAdV55) represents an emerging respiratory pathogen and causes severe pneumonia with high fatality in humans. The cellular receptors, which are essential for understanding the infection and pathogenesis of HAdV55, remain unclear. In this study, we found that HAdV55 binding and infection were sharply reduced by disrupting the interaction of viral fiber protein with human desmoglein-2 (hDSG2) but only slightly reduced by disrupting the interaction of viral fiber protein with human CD46 (hCD46). Loss-of-function studies using soluble receptors, blocking antibodies, RNA interference, and gene knockout demonstrated that hDSG2 predominantly mediated HAdV55 infection. Nonpermissive rodent cells became susceptible to HAdV55 infection when hDSG2 or hCD46 was expressed, but hDSG2 mediated more efficient HAd55 infection than hCD46. We generated two transgenic mouse lines that constitutively express either hDSG2 or hCD46. Although nontransgenic mice were resistant to HAdV55 infection, infection with HAdV55 was significantly increased in hDSG2(+/+) mice but was much less increased in hCD46(+/+) mice. Our findings demonstrate that both hDSG2 and hCD46 are able to mediate HAdV55 infection but hDSG2 plays the major roles. The hDSG2 transgenic mouse can be used as a rodent model for evaluation of HAdV55 vaccine and therapeutics. IMPORTANCE Human adenovirus type 55 (HAdV55) has recently emerged as a highly virulent respiratory pathogen and has been linked to severe and even fatal pneumonia in immunocompetent adults. However, the cellular receptors mediating the entry of HAdV55 into host cells remain unclear, which hinders the establishment of HAdV55-infected animal models and the development of antiviral approaches. In this study, we demonstrated that human desmoglein-2 (hDSG2) plays the major roles during HAdV55 infection. Human CD46 (hCD46) could also mediate the infection of HAdV55, but the efficiency was much lower than for hDSG2. We generated two transgenic mouse lines that express either hDSG2 or hCD46, both of which enabled HAd55 infection in otherwise nontransgenic mice. hDSG2 transgenic mice enabled more efficient HAdV55 infection than hCD46 transgenic mice. Our study adds to our understanding of HAdV55 infection and provides an animal model for evaluating HAdV55 vaccines and therapeutics.
- Published
- 2020
189. A CRISPR/Cas9 Generated Bovine CD46-knockout Cell Line—A Tool to Elucidate the Adaptability of Bovine Viral Diarrhea Viruses (BVDV)
- Author
-
Dirk Höper, Susanne Koethe, Kerstin Wernike, Martin Beer, and Kevin P Szillat
- Subjects
Arginine ,viruses ,Adaptation, Biological ,lcsh:QR1-502 ,knockout ,ERNS ,escape mutant ,adaptation ,Virus Replication ,lcsh:Microbiology ,Madin Darby Canine Kidney Cells ,0403 veterinary science ,Gene Knockout Techniques ,Viral Envelope Proteins ,Diarrhea Virus 2, Bovine Viral ,Asparagine ,Receptor ,CD46 ,MDBK ,Infectivity ,chemistry.chemical_classification ,0303 health sciences ,biology ,Diarrhea Virus 1, Bovine Viral ,04 agricultural and veterinary sciences ,3. Good health ,Amino acid ,Infectious Diseases ,Ribonucleoproteins ,CRISPR ,Host-Pathogen Interactions ,Receptors, Virus ,bovine viral diarrhea virus (BVDV) ,040301 veterinary sciences ,Virus ,Article ,Membrane Cofactor Protein ,03 medical and health sciences ,Dogs ,Virology ,Animals ,cell entry ,030304 developmental biology ,pestivirus ,Pestivirus ,Virus Internalization ,biology.organism_classification ,chemistry ,Amino Acid Substitution ,Cell culture ,Cattle ,CRISPR-Cas Systems ,Protein Multimerization - Abstract
Bovine viral diarrhea virus (BVDV) entry into a host cell is mediated by the interaction of the viral glycoprotein E2 with the cellular transmembrane CD46 receptor. In this study, we generated a stable Madin&ndash, Darby Bovine Kidney (MDBK) CD46-knockout cell line to study the ability of different pestivirus A and B species (BVDV-1 and -2) to escape CD46-dependent cell entry. Four different BVDV-1/2 isolates showed a clearly reduced infection rate after inoculation of the knockout cells. However, after further passaging starting from the remaining virus foci on the knockout cell line, all tested virus isolates were able to escape CD46-dependency and grew despite the lack of the entry receptor. Whole-genome sequencing of the escape-isolates suggests that the genetic basis for the observed shift in infectivity is an amino acid substitution of an uncharged (glycine/asparagine) for a charged amino acid (arginine/lysine) at position 479 in the ERNS in three of the four isolates tested. In the fourth isolate, the exchange of a cysteine at position 441 in the ERNS resulted in a loss of ERNS dimerization that is likely to influence viral cell-to-cell spread. In general, the CD46-knockout cell line is a useful tool to analyze the role of CD46 for pestivirus replication and the virus&ndash, receptor interaction.
- Published
- 2020
190. Oncolytic Virus With Attributes of Vesicular Stomatitis Virus and Measles Virus in Hepatobiliary and Pancreatic Cancers
- Author
-
Michael B. Steele, James M Bogenberger, Oumar Barro, Mitesh J. Borad, Nathan Jenks, Mansi Arora, Bolni Marius Nagalo, Yumei Zhou, Lewis R. Roberts, Camilo Breton, Alexander T. Baker, Stephen J. Russell, Kah Whye Peng, and Dan G. Duda
- Subjects
0301 basic medicine ,Cancer Research ,viruses ,PDAC, pancreatic ductal adenocarcinoma ,pancreatic cancer ,AST, aspartate aminotransferase ,DMEM, Dulbecco’s modified Eagle’s medium ,0302 clinical medicine ,PCR, polymerase chain reaction ,G, glycoprotein G ,Pharmacology (medical) ,MOI, multiplicity of infection ,H, hemagglutinin ,m, mouse ,biology ,hepatocellular carcinoma ,ELISA, enzyme-linked immunosorbent assay ,TCID50, 50% tissue culture infective dose ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,CBC, complete blood counts ,Oncology ,CPE, cytopathic effect ,eGFP, enhanced green fluorescent protein ,Vesicular stomatitis virus ,030220 oncology & carcinogenesis ,Hepatocellular carcinoma ,HBPC, hepatobiliary and pancreatic cancer ,Molecular Medicine ,wt, wild type ,vesicular stomatitis virus ,Genetically modified mouse ,VSV, vesicular stomatitis virus ,Hemagglutinin (influenza) ,F, fusion ,lcsh:RC254-282 ,Article ,hepatobiliary cancer ,Measles virus ,03 medical and health sciences ,h, human ,Pancreatic cancer ,ALT, alanine aminotransferase ,medicine ,DPBS, Dulbecco’s phosphate-buffered saline ,IFN, interferon ,HuCCT1, biliary tract cancer ,CD46 ,oncolytic viral therapy ,PE, phycoerythrin ,biology.organism_classification ,medicine.disease ,HER-2, human epidermal growth factor receptor-2 ,MV, measles virus ,Oncolytic virus ,Ed-MV, Edmonston vaccine-strain measles virus ,stomatognathic diseases ,030104 developmental biology ,NIS, sodium iodide symporter ,measles virus ,biology.protein ,Cancer research ,ATCC, American Tissue Culture Collection ,HCC, hepatocellular carcinoma - Abstract
Recombinant vesicular stomatitis virus (VSV)-fusion and hemagglutinin (FH) was developed by substituting the promiscuous VSV-G glycoprotein (G) gene in the backbone of VSV with genes encoding for the measles virus envelope proteins F and H. Hybrid VSV-FH exhibited a multifaceted mechanism of cancer-cell killing, and improved neurotolerability over parental VSV in preclinical studies. In this study, we evaluated VSV-FH in vitro and in vivo in models of hepatobiliary and pancreatic cancers. Our results indicate that high intrahepatic doses of VSV-FH did not result in any significant toxicity and were well tolerated by transgenic mice expressing the measles virus receptor CD46. Furthermore, single intratumoral treatments with VSV-FH yielded improved survival and complete tumor regressions in a proportion of mice in the Hep3B hepatocellular carcinoma model, but not in mice xenografted with BxPC3 pancreatic cancer cells. Our preliminary findings indicate that VSV-FH can induce potent oncolysis in hepatocellular and pancreatic cancer cell lines with concordant results in vivo in hepatocellular cancer and discordant in pancreatic cancer, without the VSV-mediated toxic effects previously observed in laboratory animals. Further study of VSV-FH as an oncolytic virotherapy is warranted in hepatocellular carcinoma and pancreatic cancer, to understand broader applicability and mechanisms of sensitivity and resistance., Graphical Abstract
- Published
- 2020
191. The Complement System in the Pathophysiology of Pregnancy and in Systemic Autoimmune Rheumatic Diseases During Pregnancy
- Author
-
Paola Adele Lonati, Francesco Tedesco, Pier Luigi Meroni, Laura Trespidi, and Cecilia Beatrice Chighizola
- Subjects
0301 basic medicine ,lcsh:Immunologic diseases. Allergy ,Placenta ,Immunology ,CD59 ,Review ,Autoimmune Diseases ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,systemic lupus erythematosus ,Rheumatic Diseases ,medicine ,Immunology and Allergy ,Humans ,complement ,biology ,CD46 ,business.industry ,obstetric complications ,Trophoblast ,Complement System Proteins ,Complement system ,Complement (complexity) ,Pregnancy Complications ,anti-phospholipid syndrome ,030104 developmental biology ,medicine.anatomical_structure ,biology.protein ,Female ,pregnancy ,Antibody ,business ,lcsh:RC581-607 ,030215 immunology - Abstract
The complement system plays a double role in pregnancy exerting both protective and damaging effects at placental level. Complement activation at fetal-maternal interface participates in protection against infectious agents and helps remove apoptotic and necrotic cells. Locally synthesized C1q contributes to the physiologic vascular remodeling of spiral arteries characterized by loss of smooth muscle cells and transformation into large dilated vessels. Complement activation triggered by the inflammatory process induced by embryo implantation can damage trophoblast and other decidual cells that may lead to pregnancy complications if the cells are not protected by the complement regulators CD55, CD46, and CD59 expressed on cell surface. However, uncontrolled complement activation induces placental alterations resulting in adverse pregnancy outcomes. This may occur in pathological conditions characterized by placental localization of complement fixing antibodies directed against beta2-glycoprotein 1, as in patients with anti-phospholipid syndrome, or circulating immune complexes deposited in placenta, as in patients with systemic lupus erythematosus. In other diseases, such as preeclampsia, the mechanism of complement activation responsible for complement deposits in placenta is unclear. Conflicting results have been reported on the relevance of complement assays as diagnostic and prognostic tools to assess complement involvement in pregnant patients with these disorders.
- Published
- 2020
192. The fiber knob protein of human adenovirus type 49 mediates highly efficient and promiscuous infection of cancer cell lines using a novel cell entry mechanism
- Author
-
Pierre J. Rizkallah, David K. Cole, Rosie M. Mundy, Alan L. Parker, Elise Moses, James A. Davies, Alexander T. Baker, and Gareth Marlow
- Subjects
chemistry.chemical_classification ,Phylogenetic tree ,CD46 ,virus diseases ,Biology ,Virology ,eye diseases ,Amino acid ,Oncolytic virus ,chemistry ,Immunity ,Vector (molecular biology) ,Receptor ,Tropism - Abstract
The human adenovirus (HAdV) phylogenetic tree is diverse, divided across seven species and comprising over 100 individual types. Species D HAdV are rarely isolated with low rates of pre-existing immunity, making them appealing for therapeutic applications. Several species D vectors have been developed as vaccines against infectious diseases where they induce robust immunity in pre-clinical models and early phase clinical trials. However, many aspects of the basic virology of species D HAdV, including their basic receptor usage and means of cell entry, remain understudied.Here, we investigated HAdV-D49, which previously has been studied for vaccine and vascular gene transfer applications. We generated a pseudotyped HAdV-C5 presenting the HAdV-D49 fiber knob protein (HAdV-C5/D49K). This pseudotyped vector was efficient at infecting cells devoid of all known HAdV receptors, indicating HAdV-D49 uses an unidentified cellular receptor. Conversely, a pseudotyped vector presenting the fiber knob protein of the closely related HAdV-D30 (HAdV-C5/D30K), differing in four amino acids to HAdV-D49, failed to demonstrate the same tropism. These four amino acid changes resulted in a change in isoelectric point of the knob protein, with HAdV-D49K possessing a basic apical region compared to a more acidic region in HAdV-D30K. Structurally and biologically we demonstrate that HAdV-D49 knob protein is unable to engage CD46, whilst potential interaction with CAR is extremely limited by extension of the DG loop. HAdV-C5/49K efficiently transduced cancer cell lines of pancreatic, breast, lung, oesophageal and ovarian origin, indicating it may have potential for oncolytic virotherapy applications, especially for difficult to transduce tumour types.ImportanceAdenoviruses are powerful tools experimentally and clinically. To maximise efficacy, the development of serotypes with low pre-existing levels of immunity in the population is desirable. Consequently, attention has focussed on those derived from species D, which have proven robust vaccine platforms. This widespread usage is despite limited knowledge in their basic biology and cellular tropism.We investigated the tropism of HAdV-D49, demonstrating it uses a novel cell entry mechanism that bypasses all known HAdV receptors. We demonstrate, biologically, that a pseudotyped HAdV-C5/D49K vector efficiently transduces a wide range of cell lines, including those presenting no known adenovirus receptor. Structural investigation suggests that this broad tropism is the result of a highly basic electrostatic surface potential, since a homologous pseudotyped vector with a more acidic surface potential, HAdV-C5/D30K, does not display a similar pan-tropism. Therefore, HAdV-C5/D49K may form a powerful vector for therapeutic applications capable of infecting difficult to transduce cells.
- Published
- 2020
193. gp96 Is Critical for both Human Herpesvirus 6A (HHV-6A) and HHV-6B Infections
- Author
-
Tian Tang, Xuefeng Jiang, Jingjing Ma, Jinfeng Guo, Huamin Tang, Lingyun Li, Mengyuan Xu, Kun Yao, Zhisheng Wu, Xianyi Xu, Junli Jia, and Benshun Hu
- Subjects
Herpesvirus 6, Human ,T-Lymphocytes ,viruses ,T cell ,Receptor expression ,Primary Cell Culture ,Immunology ,Cell ,Roseolovirus Infections ,Biology ,Microbiology ,Cell Line ,03 medical and health sciences ,0302 clinical medicine ,Viral Envelope Proteins ,Viral entry ,Virology ,medicine ,Humans ,CD134 ,Tropism ,030304 developmental biology ,0303 health sciences ,Membrane Glycoproteins ,CD46 ,virus diseases ,Virus Internalization ,biochemical phenomena, metabolism, and nutrition ,Fetal Blood ,Entry into host ,Virus-Cell Interactions ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Insect Science ,Protein Binding - Abstract
Human herpesviruses 6A and 6B (HHV-6A and HHV-6B, respectively) are two virus species in the betaherpesvirus subfamily that exhibit T cell tropism. CD46 and CD134 are the cellular receptors for HHV-6A and HHV-6B, respectively. Interestingly, the efficiency of HHV-6A/6B entry is different among different types of target cells despite similar receptor expression levels on these cells. Here, we found that the cellular factor gp96 (also known as glucose-regulated protein 94 [GRP94]) is expressed on the cell surface and interacts with viral glycoprotein Q1 (gQ1) during virus entry. gp96 cell surface expression levels are associated with the efficiency of HHV-6A and HHV-6B entry into target cells. Both loss-of-function and gain-of-function experiments indicated that gp96 plays an important role in HHV-6 infection. Our findings provide new insight into the HHV-6 entry process and might suggest novel therapeutic targets for HHV-6 infection. IMPORTANCE Although new clinical importance has been revealed for human herpesviruses 6A (HHV-6A) and 6B, much is still unknown about the life cycles of these viruses in target cells. We identified a novel cellular factor, gp96, that is critical for both HHV-6A and -6B entry into host cells. As gp96 can function as an adjuvant in vaccine development for both infectious agents and cancers, it can be a potential therapeutic target for infection by these two viruses.
- Published
- 2020
194. Complement activation in sickle cell disease: Dependence on cell density, hemolysis and modulation by hydroxyurea therapy
- Author
-
Lubka T. Roumenina, Philippe Chadebech, Gwellaouen Bodivit, Paula Vieira‐Martins, Anne Grunenwald, Idris Boudhabhay, Victoria Poillerat, Sadaf Pakdaman, Laurent Kiger, Alicia Jouard, Etienne Audureau, France Pirenne, Frédéric Galactéros, Véronique Frémeaux‐Bacchi, Pablo Bartolucci, Centre de Recherche des Cordeliers (CRC (UMR_S_1138 / U1138)), École pratique des hautes études (EPHE), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU)-Université de Paris (UP), Etablissement Français du Sang [Île-de-France Mondor], IMRB - 'Transfusion et Maladies du Globule Rouge' [Créteil] (U955 Inserm - UPEC), Institut Mondor de Recherche Biomédicale (IMRB), Institut National de la Santé et de la Recherche Médicale (INSERM)-IFR10-Université Paris-Est Créteil Val-de-Marne - Paris 12 (UPEC UP12)-Institut National de la Santé et de la Recherche Médicale (INSERM)-IFR10-Université Paris-Est Créteil Val-de-Marne - Paris 12 (UPEC UP12), Hôpital Européen Georges Pompidou [APHP] (HEGP), Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpitaux Universitaires Paris Ouest - Hôpitaux Universitaires Île de France Ouest (HUPO), Hôpital Henri Mondor, École pratique des hautes études (EPHE)-Université Paris Diderot - Paris 7 (UPD7)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU), Institut National de la Santé et de la Recherche Médicale (INSERM)-IFR10-Université Paris-Est Créteil Val-de-Marne - Paris 12 (UPEC UP12), Substitut du sang et pathologie moléculaire du globule rouge, Institut National de la Santé et de la Recherche Médicale (INSERM), Assistance publique - Hôpitaux de Paris (AP-HP) (APHP)-Hôpital Henri Mondor-Université Paris-Est Créteil Val-de-Marne - Paris 12 (UPEC UP12), Unité des maladies génétiques du globule rouge, Assistance publique - Hôpitaux de Paris (AP-HP) (APHP)-Hôpital Henri Mondor-Centre de référence des syndromes drépanocytaires majeurs-Université Paris-Est Créteil Val-de-Marne - Paris 12 (UPEC UP12), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Université Paris Diderot - Paris 7 (UPD7)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris-Est Créteil Val-de-Marne - Paris 12 (UPEC UP12)-IFR10, Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpital Henri Mondor-Université Paris-Est Créteil Val-de-Marne - Paris 12 (UPEC UP12), Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpital Henri Mondor-Centre de référence des syndromes drépanocytaires majeurs-Université Paris-Est Créteil Val-de-Marne - Paris 12 (UPEC UP12), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU)-Université Paris Cité (UPCité), and CHADEBECH, Philippe
- Subjects
[SDV.MHEP.HEM] Life Sciences [q-bio]/Human health and pathology/Hematology ,Adult ,medicine.medical_specialty ,Adolescent ,Cell Count ,CD59 ,Anemia, Sickle Cell ,[SDV.GEN.GH] Life Sciences [q-bio]/Genetics/Human genetics ,[SDV.IMM.II]Life Sciences [q-bio]/Immunology/Innate immunity ,Hemolysis ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,Downregulation and upregulation ,Internal medicine ,hemic and lymphatic diseases ,medicine ,Humans ,Hydroxyurea ,complement ,[SDV.BBM.BC]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Biochemistry [q-bio.BM] ,heme ,[SDV.BBM.BC] Life Sciences [q-bio]/Biochemistry, Molecular Biology/Biochemistry [q-bio.BM] ,Complement Activation ,030304 developmental biology ,Whole blood ,0303 health sciences ,Innate immune system ,Chemistry ,CD46 ,Sickle cell disease ,Hemopexin ,[SDV.MHEP.HEM]Life Sciences [q-bio]/Human health and pathology/Hematology ,Hematology ,Middle Aged ,medicine.disease ,sC5b-9 ,3. Good health ,Complement system ,Endocrinology ,[SDV.GEN.GH]Life Sciences [q-bio]/Genetics/Human genetics ,[SDV.IMM.IA]Life Sciences [q-bio]/Immunology/Adaptive immunology ,[SDV.IMM.IA] Life Sciences [q-bio]/Immunology/Adaptive immunology ,Female ,030215 immunology - Abstract
International audience; The complement system is an innate immune defense cascade that can cause tissue damage when inappropriately activated. Evidence for complement over activation has been reported in small cohorts of patients with sickle cell disease (SCD). However, the mechanism governing complement activation in SCD has not been elucidated. Here, we observe that the plasma concentration of sC5b-9, a reliable marker for terminal complement activation, is increased at steady state in 61% of untreated SCD patients. We show that greater complement activation in vitro is promoted by SCD erythrocytes compared to normal ones, although no significant differences were observed in the regulatory proteins CD35, CD55, and CD59 in whole blood. Complement activation is positively correlated with the percentage of dense sickle cells (DRBCs). The expression levels of CD35, CD55, and CD59 are reduced in DRBCs, suggesting inefficient regulation when cell density increases. Moreover, the surface expression of the complement regulator CD46 on granulocytes was inversely correlated with the plasma sC5b-9. We also show increased complement deposition in cultured human endothelial cells incubated with SCD serum, which is diminished by the addition of the heme scavenger hemopexin. Treatment of SCD patients with hydroxyurea produces substantial reductions in complement activation, measured by sC5b-9 concentration and upregulation of CD46, as well as decreased complement activation on RBCs in vitro. In conclusion, complement over activation is a common pathogenic event in SCD that is associated with formation of DRBCs and hemolysis. And, it affects red cells, leukocytes and endothelial cells. This complement over activation is partly alleviated by hydroxyurea therapy.
- Published
- 2020
195. The landscape of alternative splicing in HIV-1 infected CD4 T-cells
- Author
-
Vicente Planelles, Yue Zheng, Younghee Lee, Seonggyun Han, and Seyoun Byun
- Subjects
CD4-Positive T-Lymphocytes ,Genetic Markers ,lcsh:Internal medicine ,Candidate gene ,Cyclin T1 ,lcsh:QH426-470 ,HIV Infections ,Major histocompatibility complex ,Virus Replication ,Exon ,Viral life cycle ,Genetics ,Transcriptional regulation ,Humans ,Gene Regulatory Networks ,lcsh:RC31-1245 ,Gene ,CD46 ,Genetics (clinical) ,CCNT1 ,biology ,Sequence Analysis, RNA ,Research ,Alternative splicing ,Computational Biology ,Exons ,lcsh:Genetics ,Alternative Splicing ,CD4 T-cell ,biology.protein ,HIV-1 - Abstract
Background Elucidating molecular mechanisms that are altered during HIV-1 infection may provide a better understanding of the HIV-1 life cycle and how it interacts with infected T-cells. One such mechanism is alternative splicing (AS), which has been studied for HIV-1 itself, but no systematic analysis has yet been performed on infected T-cells. We hypothesized that AS patterns in infected T-cells may illuminate the molecular mechanisms underlying HIV-1 infection and identify candidate molecular markers for specifically targeting infected T-cells. Methods We downloaded previously published raw RNA-seq data obtained from HIV-1 infected and non-infected T-cells. We estimated percent spliced in (PSI) levels for each AS exon, then identified differential AS events in the infected cells (FDR 0.1). We performed functional gene set enrichment analysis on the genes with differentially expressed AS exons to identify their functional roles. In addition, we used RT-PCR to validate differential alternative splicing events in cyclin T1 (CCNT1) as a case study. Results We identified 427 candidate genes with differentially expressed AS exons in infected T-cells, including 20 genes related to cell surface, 35 to kinases, and 121 to immune-related genes. In addition, protein-protein interaction analysis identified six essential subnetworks related to the viral life cycle, including Transcriptional regulation by TP53, Class I MHC mediated antigen, G2/M transition, and late phase of HIV life cycle. CCNT1 exon 7 was more frequently skipped in infected T-cells, leading to loss of the key Cyclin_N motif and affecting HIV-1 transcriptional elongation. Conclusions Our findings may provide new insight into systemic host AS regulation under HIV-1 infection and may provide useful initial candidates for the discovery of new markers for specifically targeting infected T-cells.
- Published
- 2020
196. Ayu (Plecoglossus altivelis) CD46 isoforms protect the cells from autologous complement attack
- Author
-
Jiong Chen, Wen-Jing Ma, and Yu-Hong Shi
- Subjects
0301 basic medicine ,Gene isoform ,Fish Proteins ,Aquatic Science ,Biology ,law.invention ,Membrane Cofactor Protein ,03 medical and health sciences ,Fish Diseases ,law ,Environmental Chemistry ,Animals ,Amino Acid Sequence ,Phylogeny ,CD46 ,Gene Expression Profiling ,Alternative splicing ,Immunity ,04 agricultural and veterinary sciences ,General Medicine ,Molecular biology ,Transmembrane protein ,Complement system ,Blot ,030104 developmental biology ,Gene Expression Regulation ,Osmeriformes ,040102 fisheries ,Recombinant DNA ,0401 agriculture, forestry, and fisheries ,Plecoglossus altivelis ,Sequence Alignment - Abstract
CD46 is an important immune regulatory receptor with multiple functions. However, studies on the function of teleost CD46, especially the different CD46 isoforms are limited. In this study, we identified three membrane cofactor protein (MCP, CD46) gene isoforms from ayu (Plecoglossus altivelis) and tentatively named as PaCD46 isoforms. PaCD46 isoforms were generated by alternative splicing and all consisted of four conserved short consensus repeats (SCRs), and the variable serine-threonine-proline-rich domain, transmembrane hydrophobic domain, and cytoplasmic tail. Phylogenetic analysis showed that the isoforms clustered together with other fish CD46 and then with higher animal CD46. Western blotting analysis of peripheral blood mononuclear cells (PBMC) revealed three bands, all of which had much larger molecular weights than the theoretical values of the three PaCD46 isoforms. Moreover, three PaCD46 isoforms were individually expressed on HEK293 cells, and Western blotting showed the similar band profile to that of PBMC. The recombinant extracellular domain of the PaCD46 isoforms, obtained by expression in Pichia pastoris, significantly reduced hemolysis activity of ayu sera. Furthermore, each of the three PaCD46 isoforms respectively protected the HEK293 cells expressing the isoform. The isoforms were also identified for their protection of autologous PBMC from complement activation. These results provided the first evidence that PaCD46 isoforms may be complement regulatory proteins to prevent complement-induced damage to self-tissue.
- Published
- 2020
197. Sex Differences in Clinical Presentation and Outcomes among Patients with Complement-Gene-Variant-Mediated Thrombotic Microangiopathy
- Author
-
Natalja Haninger-Vacariu, Leah Charlotte Piggott, Nóra Garam, Christof Aigner, Martina Gaggl, Alice Schmidt, Gere Sunder-Plassmann, Zoltán Prohászka, Dorottya Csuka, Raute Sunder-Plassmann, and Renate Kain
- Subjects
medicine.medical_specialty ,Thrombotic microangiopathy ,medicine.medical_treatment ,030232 urology & nephrology ,lcsh:Medicine ,Renal function ,Article ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,gender ,Medicine ,sex ,Renal replacement therapy ,orphan disease ,030304 developmental biology ,0303 health sciences ,business.industry ,CD46 ,lcsh:R ,Haplotype ,genetic renal disease ,General Medicine ,Eculizumab ,medicine.disease ,clinical nephrology ,thrombotic microangiopathy ,Factor H ,Cohort ,hemolytic uremic syndrome ,business ,medicine.drug - Abstract
Sex differences among patients with complement-gene-variant-mediated thrombotic microangiopathy (cTMA) are not well established. We examined demographic and clinical data from female and male patients with a history of cTMA enrolled in the Vienna thrombotic microangiopathy (TMA) cohort. Follow-up was three years after first presentation with cTMA. In this single-center study, we identified 51 patients with a first manifestation of cTMA between 1981 and 2019, 63% were female (p = 0.09). The median age at diagnosis did not differ between females and males. There was also no disparity between the sexes with regard to renal function or the need for renal replacement therapy at presentation. Furthermore, we observed similar use of plasma or eculizumab therapy and a comparable evolution of renal function of female and male patients. More females showed risk haplotypes of complement factor H (CFH) and CD46 (97% vs. 68%, p = 0.01), but there was no difference in the prevalence of rare pathogenic variants in complement-associated genes with regard to sex. In conclusion, the majority of cTMA patients enrolled in the Vienna TMA cohort were female. Clinical presentation and renal function did not differ between the sexes, but females more frequently presented with cTMA risk haplotypes.
- Published
- 2020
198. Selective germline genome edited pigs and their long immune tolerance in Non Human Primates
- Author
-
Zha G, Juan-Nan Chen, Y. Zhang, Sixiong Lin, Lijin Zou, Yu H, Zhong G, Yu Y, Xuenong Zou, Menghan Gao, Guangqian Zhou, Guo-Liang Wang, Jun Wang, Kenli Li, ying He, Dinghui Liu, Lei W, Linxian Li, and Yuanlin Zeng
- Subjects
medicine.medical_specialty ,CD46 ,Somatic cell ,Xenotransplantation ,medicine.medical_treatment ,Biology ,Germline ,Organ transplantation ,Natural killer cell ,Genetically modified organism ,Immune tolerance ,medicine.anatomical_structure ,Immunology ,medicine - Abstract
Organ transplantation is the only curative treatment for patients with terminal organ failure, however, there is a worldwide organ shortage. Genetically modified pig organs and tissues have become an attractive and practical alternative solution for the severe organ shortage, which has been made possible by significant progress in xenotransplantation in recent years. The past several decades witnessed an expanding list of genetically engineered pigs due to technology advancements, however, the necessary combination of genetic modifications in pig for human organ xenotransplantation has not been determined. In the current study, we created a selective germline genome edited pig (SGGEP). The first triple xenoantigens (GGTA, B4GAL, and CAMH) knockout somatic cells were generated to serve as a prototype cells and then human proteins were expressed in the xenoantigen knockout cells, which include human complement system negative regulatory proteins (CD46, CD55, and CD59); human coagulation system negative regulatory proteins thrombomodulin (THBD); tissue factor pathway inhibitor (TFPI); CD39; macrophage negative regulatory proteins (human CD47); and natural killer cell negative regulatory human HLA-E. After the successful establishment of SGGEP by the nuclear tranfer, we engrafted SGGEP skin to NHP, up to 25 days graft survival without immunosuppressive drugs was observed. Because a pig skin graft does not impact the success of a subsequent allograft or autograft or vice versa, thus our SGGEP could have a great potential for clinical value to save severe and large area burn patients and the other human organ failure. Therefore, this combination of specific gene modifications is a major milestone and provides proof of concept to initiate investigator-initiated clinical trials (IITs) in severe burn patients with defined processes and governance measures in place and the other clinical application.
- Published
- 2020
- Full Text
- View/download PDF
199. Viable pigs after simultaneous inactivation of porcine MHC class I and three xenoreactive antigen genes GGTA1, CMAH and B4GALNT2
- Author
-
Robert Rieben, Wei Liang, Beate Rieblinger, Angelika Schnieke, Reinhard Schwinzer, Eckhard Wolf, Julia Zuber, Riccardo Sfriso, Konrad Fischer, Alexander Kind, Mayuko Kurome, Bernhard Klinger, Andrea Fischer, Valeri Zakhartchenko, Rabea Hein, Barbara Kessler, and Krzysztof Flisikowski
- Subjects
Graft Rejection ,0301 basic medicine ,Swine ,Xenotransplantation ,medicine.medical_treatment ,Transplantation, Heterologous ,Immunology ,Antibodies, Heterophile ,610 Medicine & health ,Human leukocyte antigen ,030230 surgery ,Mixed Function Oxygenases ,03 medical and health sciences ,0302 clinical medicine ,Antigen ,HLA Antigens ,MHC class I ,medicine ,Animals ,Humans ,Cells, Cultured ,Gene knockout ,Transplantation ,biology ,CD46 ,Histocompatibility Antigens Class I ,Complement System Proteins ,Galactosyltransferases ,Kidney Transplantation ,Molecular biology ,ddc ,Complement system ,030104 developmental biology ,biology.protein ,Heterografts ,N-Acetylgalactosaminyltransferases ,570 Life sciences ,CRISPR-Cas Systems ,Antibody - Abstract
BACKGROUND Cell surface carbohydrate antigens play a major role in the rejection of porcine xenografts. The most important for human recipients are α-1,3 Gal (Galactose-alpha-1,3-galactose) causing hyperacute rejection, also Neu5Gc (N-glycolylneuraminic acid) and Sd(a) blood group antigens both of which are likely to elicit acute vascular rejection given the known human immune status. Porcine cells with knockouts of the three genes responsible, GGTA1, CMAH and B4GALNT2, revealed minimal xenoreactive antibody binding after incubation with human serum. However, human leucocyte antigen (HLA) antibodies cross-reacted with swine leucocyte antigen class I (SLA-I). We previously demonstrated efficient generation of pigs with multiple xeno-transgenes placed at a single genomic locus. Here we wished to assess whether key xenoreactive antigen genes can be simultaneously inactivated and if combination with the multi-transgenic background further reduces antibody deposition and complement activation. METHODS Multiplex CRISPR/Cas9 gene editing and somatic cell nuclear transfer were used to generate pigs carrying functional knockouts of GGTA1, CMAH, B4GALNT2 and SLA class I. Fibroblasts derived from one- to four-fold knockout animals, and from multi-transgenic cells (human CD46, CD55, CD59, HO1 and A20) with the four-fold knockout were used to examine the effects on human IgG and IgM binding or complement activation in vitro. RESULTS Pigs were generated carrying four-fold knockouts of important xenoreactive genes. In vitro assays revealed that combination of all four gene knockouts reduced human IgG and IgM binding to porcine kidney cells more effectively than single or double knockouts. The multi-transgenic background combined with GGTA1 knockout alone reduced C3b/c and C4b/c complement activation to such an extent that further knockouts had no significant additional effect. CONCLUSION We showed that pigs carrying several xenoprotective transgenes and knockouts of xenoreactive antigens can be readily generated and these modifications will have significant effects on xenograft survival.
- Published
- 2020
200. Is the atypical hemolytic uremic syndrome risk polymorphism in Membrane Cofactor Protein MCPggaac relevant in kidney transplantation? A case report
- Author
-
Emilia Arjona, Santiago Rodríguez de Córdoba, Ana Sánchez-Moreno, Francisco de la Cerda, Rafael Bedoya, Julia Fijo, Adela Rodríguez-Barba, Sánchez-Moreno, Ana [0000-0003-0684-7598], Rodríguez-Barba, Adela [0000-0003-4382-3085], Arjona, Emilia [0000-0002-0753-3657], Rodríguez de Córdoba, Santiago [0000-0001-6401-1874], Sánchez-Moreno, Ana, Rodríguez-Barba, Adela, Arjona, Emilia, and Rodríguez de Córdoba, Santiago
- Subjects
medicine.medical_specialty ,Complement factor B ,030232 urology & nephrology ,030230 surgery ,urologic and male genital diseases ,Extrarenal involvement ,Risk Assessment ,Gastroenterology ,Membrane Cofactor Protein ,Kidney transplantation ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,hemic and lymphatic diseases ,Atypical hemolytic uremic syndrome ,medicine ,Genetic predisposition ,Membrane cofactor protein ,Humans ,Hemolytic uremic syndrome ,Atypical Hemolytic Uremic Syndrome ,Transplantation ,Polymorphism, Genetic ,CD46 ,business.industry ,Infant ,Eculizumab ,medicine.disease ,Kidney Transplantation ,Pedigree ,Pediatrics, Perinatology and Child Health ,Female ,business ,medicine.drug ,Kidney disease ,Rare disease ,Polymorphism MCP - Abstract
4 p.-1 fig., aHUS is a rare disease characterized by episodes of TMA that frequently progresses to CKD and often recurs after KT. The most frequent cause of aHUS is defective regulation of complement activation because of genetic anomalies. Eculizumab interrupts the process of TMA and improves renal function. We describe one female patient with aHUS who debuted in 2005 at 3-mo-old with extrarenal manifestations and progressed to end-stage kidney disease (ESKD) within a year. Her family history included several affected members with similar bad outcomes. Our patient carries a strong aHUS genetic predisposition consisting in a pathogenic gain-of-function mutation in complement factor B concurrent with the MCP aHUS risk haplotype MCPggaac. She received a kidney transplant in 2011 without eculizumab prophylaxis.The graft, which was negative for the MCPggaac risk haplotype, had an unexpected excellent evolution without aHUS recurrence. Different retrospective studies have shown that the risk of aHUS recurrence after KT correlates well with the genetic load of aHUS risk factors. Knowing important contribution of the MCPggaac risk haplotype to the risk of developing aHUS in Factor B mutations carriers, we speculate whether the absence of this polymorphism in the graft that our patient received may have decreased the risk of aHUS recurrence after KT.
- Published
- 2020
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.