Bioinformatics Resource Centers (BRCs) for Infectious Diseases

The NIAID-funded Bioinformatics Resource Centers provide data-driven, production-level, sustainable computational platforms to enable sharing and access to data, portable computational tools, and standards that support interoperability for the infectious diseases research community.

Laboratory of Immune System Biology

Ronald N. Germain, M.D., Ph.D., Chief

The major research activities of Laboratory of Immune System Biology (LISB) are focused on the basic genetics, molecular biology, and cell biology of the immune system, as well as on human disease informed by these more basic studies. How dysregulation of the immune system results in immunodeficiencies, autoimmunity, inflammation, allergy, chronic infections, and lymphoproliferative diseases and what strategies might be valuable for therapeutic or vaccine development related to these conditions as well as cancer are important topics of interest, as is the behavior of the meta-organism (the combination of the host and the commensal microbiota).

A key aspect of LISB research is the development and application of systems and quantitative approaches to dissect how the molecules, cells, and commensals studied by LISB investigators together shape emergent immune system behavior. Modern technology now allows the analysis of immune responses and host-pathogen interactions at a global level, across scales ranging from molecular interactions to intracellular signaling and gene regulatory networks to individual cell behavior to the functioning of a tissue, an organ, and the whole organism. The challenge is to organize and integrate this information to extract biological insights that can help enhance our understanding of how the immune system operates in health or disease or how pathogens affect their hosts. To move towards these goals, LISB scientists seek to collect detailed quantitative as well as qualitative data on the state, organization, and operation of the immune system to develop computational models (mechanistic, statistical, and machine-learning) that can be used to predict the behavior of a complex biological system, uncover the components involved, and help explain the mechanistic basis for physiological and pathological responses to infection or vaccination or to design new therapies or vaccines.

Achieving this goal requires an interdisciplinary effort, and LISB is designed to address this challenge. Although it is composed of independent laboratories, the LISB is intended to operate in many of its research efforts as an integrated group of scientists and support staff. Although it has been established within NIAID, it is expected to play a major role in fostering the growth of immunology and systems biology efforts across the National Institutes of Health (NIH) through its development of software tools for complex systems modeling, new experimental paradigms, advanced imaging modalities, high-throughput screening efforts, and novel approaches to microbiome analysis. LISB members are involved in an extensive web of formal and informal interactions with other intramural NIH scientists and with extramural groups in the United States and abroad that have a common interest in a systems approach to biology in general and immunology in particular.

View all Division of Intramural Research laboratories

Major Areas of Research

  • Quantitative systems-level studies of humans and of animal models
  • Development, differentiation, and plasticity of immune cells, including T cells, B cells, and innate lymphoid cells
  • Transcriptional and post-transcriptional regulation of lymphocyte differentiation and function
  • Regulation of primary and secondary immune responses
  • Mechanisms of pattern recognition and antigen receptor signaling and transcriptional control
  • Gene expression profiling, high-content imaging of immune signaling pathways, RNAi and CRISPR/Cas9 for the discovery of pathway components, high-throughput proteomic and genomic analysis
  • Translational immunogenomics: integrative analysis of the human immune system’s response to vaccines, medications, and disease states, in vitro and in vivo
  • Detection and analysis of gene defects and pathogenetic mechanisms in human inborn errors of immunity
  • Single-cell biology: quantitative and functional analysis of cellular heterogeneity and stochastic gene expression
  • Advanced dynamic and static multiplex imaging of mouse and human tissues
  • RNA-binding protein and miRNA-mediated regulation of immune cells
  • Host-pathogen interactions
  • Immune-microbiota interactions
  • Role of nutrition on the immune system
  • Tissue-specific immunity
  • Fetal hematopoietic stem cells
  • Programmed cell death and autophagy
  • Biology of regulatory T cells and their role in autoimmunity and chronic infection
  • Induction of T-cell tolerance and treatment of autoimmunity
  • Structural and functional studies of macromolecules involved in antigen processing and presentation, adaptive and innate immune recognition, and viral immunoevasion
  • Detection and analysis of gene defects pathogenetic mechanisms in human inborn errors of immunity

See a playlist of videos of LISB research.

Simmune Project

Simmune is a suite of software tools that guides the user through the multiple hierarchical scales of cellular behavior, facilitating the generation of comprehensive models. It was originally created to simulate immunological phenomena—hence its name, Simmune—but it is applicable to a very broad class of cell biological models.


Read more about the Simmune Project

Laboratory of Infectious Diseases

Jeffrey I. Cohen, M.D., Chief

Established in 1942, the Laboratory of Infectious Diseases (LID) has a long history of vaccine development and identification of new agents of viral diseases. LID is noted for undertaking high-risk, high-re​ward programs that require extraordinary time and resource commitments, such as programs to develop vaccines for viral hepatitis, severe childhood respiratory diseases, viral gastroenteritis, flaviviruses, and herpesviruses.

Clinical studies complement LID’s major areas of research, including testing candidate vaccines in clinical trials, human challenge studies with influenza and respiratory syncytial virus to study pathogenesis and immune correlates for protection against these viruses, and studies of severe virus infections in persons without known immune deficiency.

View all Division of Intramural Research laboratories

Examples of LID Accomplishments

  • The first approved hepatitis A vaccine in the US (Havrix) and a monoclonal antibody to respiratory syncytial virus (RSV) (Synagis).
  • A live, attenuated influenza vaccine, FluMist (Medimmune) developed by LID with participation of extramural NIAID.
  • Vaccines against potential pandemic influenza strains in collaboration with Medimmune tested in Phase I clinical trials.
  • The first approved rotavirus vaccine in the US (RotaShield). 
  • A bovine-human reassortant rotavirus vaccine, RotaSIIL used in India. A USPTO Patents for Humanity award to NIAID for RotaSIIL in 2018.
  • A recombinant live, attenuated RSV vaccine shown to be safe and immunogenic in 1- to 2-month-old infants and continuing in clinical trials.
  • A hepatitis E vaccine, licensed to GlaxoSmithKline, proved highly effective in preventing hepatitis E in a field trial in Nepal.
  • A vaccine candidate for West Nile virus was shown to be safe and immunogenic in young adults and in the elderly.
  • A tetravalent live, attenuated dengue virus vaccine in a Phase III clinical trial and licensed throughout the world.
  • A live, attenuated PIV3 vaccine passed Phase II safety trials in infants; PIV1 and PIV2 vaccine candidates generated.
  • A diagnostic for norovirus, RIDASCREEN, licensed.
  • Human volunteer influenza and respiratory syncytial virus challenge models developed for testing vaccines, immunotherapeutics, and antivirals.

Former Senior Investigators

Listing of former Senior Investigators with the Laboratory of Infectious Diseases


Former Senior Investigators

Centers for Research on Structural Biology of Infectious Diseases (CRSTAL-ID)—Resources

The Centers for Research on Structural Biology of Infectious Diseases (CRSTAL-ID) provide the research community with: 3-D protein structures and protein-ligand complexes; Sequence-verified clones and peptides; Services that deliver requested 3-D structure determination; and Molecular screening of proteins in complex with inhibitors, cofactors and substrate analogs

Early Phase Clinical Trial Units

The Early Phase Clinical Trial Units (EPCTUs) support the design, development, implementation, and conduct of Phase 0 to Phase 2 clinical trials, including proof-of-concept studies in healthy, special, and disease-specific populations, carried out in an expeditious and efficient manner, aided by bioanalysis when necessary, against viral, bacterial, parasitic, and fungal pathogens and emerging and re-emerging infectious diseases. The candidate products assessed through the EPCTUs may include licensed or investigational products such as small molecules, monoclonal antibodies, i

Genomic Centers for Infectious Diseases (GCID) Resources

The GCID use, develop and improve innovative applications of genomic technologies, such as DNA and RNA sequencing and metagenomics, and provide rapid and cost-efficient production of high-quality genome sequences of microorganisms, invertebrate vectors of infectious diseases, and hosts and host microbiomes.

Joseph Marcotrigiano, Ph.D.

Chief, Structural Virology Section

Major Areas of Research

  • Explore the mechanism of entry and replication of RNA viruses
  • Understand how the cell distinguishes self from non-self 
  • Characterize the immune response to RNA virus infection
  • Contribute to the development of novel therapies to combat infection and spread of RNA viruses
  • Develop novel methods for the recombinant production of challenging proteins in mammalian cells. 

Program Description

Our laboratory is committed to understanding how positive-sense RNA viruses enter human host cells, replicate, and evade the immune response. Toward this overarching goal we are (1) determining how hepatitis C virus (HCV) glycoproteins with host cell receptors and broadly neutralizing antibodies, (2) exploring the mechanisms of viral polyprotein processing, and (3) examining how the innate immune system distinguishes self versus viral RNAs. Our long-term goal is to discover and develop an effective HCV vaccine, novel antiviral drugs, and RIG-I immunomodulators for use as broad-based antiviral agents.

HCV represents an important public health problem, infecting an estimated 2% of the world’s population, approximately 150 million people. There is no vaccine against HCV and it is estimated that an additional 3-4 million individuals become newly infected each year worldwide. Several direct acting antivirals for HCV have recently been FDA approved; however, there is still a need for an HCV vaccine as these drugs are prohibitively expensive. HCV is an enveloped virus with an outer shell composed of many copies of two glycoproteins, E1 and E2. Mechanistic studies aimed at providing a detailed understanding of HCV entry have lagged behind due to an inability to produce properly folded HCV glycoproteins. To circumvent this obstacle, my laboratory has developed a cost-, labor-, and time-efficient method for the large-scale production of recombinant glycoproteins in mammalian cell lines with native, post-translational modifications. This production system enabled the first determination of the core domain of HCV E2 structure in complex with an Fab (Khan et al. Nature 2014). This structure revealed that HCV E2 does not share any similarity to other viral glycoproteins, including those from closely related viruses, suggesting that HCV may use a novel entry mechanism.

Numerous viruses, many of which severely impact human health around the globe (e.g. HIV, Zika virus, Dengue virus, West Nile virus, chikungunya virus, and SARS virus), utilize a gene expression mechanism wherein one gene encodes a single polyprotein that is post-translationally cleaved into individual proteins. Proteolytic processing occurs in a highly regulated and coordinated manner with intermediates having distinct functions and playing important roles during viral infection. Remarkably, viral proteases found within the polyprotein are capable of performing multiple cleavages that can be separated by long distances in linear sequence. Despite the commonality of polyprotein processing as a means of regulating the viral life cycle, very little is known about the higher order complexes of replication proteins post-processing, and even less about the structural properties of the precursor forms. Much of the existing biochemical and biophysical information on viral proteins stems from structural results of individual domains or mature forms of the viral proteins. To fully understand the biology and the regulation behind each step of the viral life cycle, it is absolutely necessary to examine the changes in the properties of viral proteins before and after polyprotein processing. We have determined the structure of the precleavage form of a portion of the Alphavirus replication machinery, helping to illuminate these critical steps in the virus life cycle (Shin et al. PNAS 2012). Our results have provided unprecedented insights into viral polyprotein processing and pathogenesis, which may be applicable to other important human viruses that undergo polyprotein processing.

The innate immune system is an evolutionarily ancient component of the host defense mechanisms. The innate immune receptors can efficiently distinguish general features of pathogens in a background of related self-molecules. The RIG-I (retinoic-acid-inducible gene-I) like receptors (RLR) consist of three cytoplasmic proteins (RIG-I, MDA5 and LGP2) that detect the presence of viral RNA in infected cells. A major goal of our research is to understand the mechanism of self versus non-self recognition and immune signaling actions of the RLR family receptors. To understand the synergy between the various domains of RIG-I for RNA binding and activation, my laboratory determined the structure of the RNA binding domains of human RIG-I in complex with dsRNA and an ATP analogue (Jiang et al. Nature, 2011). Recently, we established that the 2’O-methyl group on the first nucleotide adjacent to the 7-methyl guanosine cap is an important self versus non-self marker for RIG-I and demonstrated that RNAs with only 7-methyl guanosine caps are PAMPs (Devarkar et al. PNAS 2016).

Biography

Dr. Joseph Marcotrigiano pursued graduate studies at Rockefeller University in the laboratory of Dr. Stephen K. Burley, determining the first structures of proteins involved in eukaryotic translation initiation. After receiving a Ph.D., Dr. Marcotrigiano became a Merck Fellow of the Life Sciences Research Foundation at the Center for the Study of Hepatitis C under the direction of Dr. Charles Rice. As a postdoctoral fellow, he determined the structures of HCV nonstructural protein (NS) 5A and NS2. In 2007, he began an independent tenure-track position at the Center for Advanced Biotechnology and Medicine at Rutgers University and was awarded tenure in July 2013. In September 2016, he was selected as a Howard Hughes Medical Institute Faculty Scholar. Dr. Marcotrigiano became chief of the Structural Virology Section in the Laboratory of Infectious Diseases in January 2017.

Research Group

Jennifer Casino-Matos
Chen Wang
Yuanyuan Wang

Selected Publications

Devarkar S, Wang C, Miller MT, Ramanathan A, Jiang F, Khan AG, Patel S, Marcotrigiano J. Structural basis for m7G recognition and 2’-O-methyl discrimination in capped RNAs by the innate immune receptor RIG-I. Proceedings of the National Academy of Sciences USA. 2016 Jan 19;113(3):596-601.

Khan A, Whidby J, Miller M, Scarborough H, Zatorski A, Cygan A, Price A, Yost S, Bohannon C, Jacob J, Grakoui A, Marcotrigiano J. Structure of the core ectodomain of the hepatitis C virus envelope glycoprotein 2. Nature. 2014 May 15;509(7500):381-4.

Shin G, Yost S, Miller M, Elrod E, Grakoui A, Marcotrigiano J. Structural and Functional Insights into Alphavirus Polyprotein Processing and Pathogenesis. Proceedings of the National Academy of Sciences (PNAS) USA. 2012 Oct 9;109(41):16534-9.

Jiang F, Ramanathan A, Miller MT, Tang G-Q, Gale M Jr., Patel SS, Marcotrigiano J. Structural basis of RNA recognition and activation by innate immune receptor RIG-INature. 2011 Sep 25;479(7373):423-7.

Whidby J, Mateu G, Scarborough H, Demeler B, Grakoui A, Marcotrigiano J. Blocking hepatitis C virus infection with recombinant form of envelope protein 2 ectodomain. Journal of Virology. 2009 Nov;83(21):11078-89.

Saito T, Owen DM, Jiang F, Marcotrigiano J, Gale M Jr. Innate immunity induced by composition-dependent RIG-I recognition of hepatitis C virus RNA. Nature. 2008 Jul 24;454(7203):523-7.

Section or Unit Name
Structural Virology Section
First Name
Joseph
Last Name
Marcotrigiano
Suffix
Ph.D.
Exclude from directory
Off
Section/Unit: Year Established
Section/Unit: Location
This Researcher/Clinician’s Person Page
Program Description

Our laboratory is committed to understanding how positive-sense RNA viruses enter human host cells, replicate, and evade the immune response. Toward this overarching goal we are (1) determining how hepatitis C virus (HCV) glycoproteins with host cell receptors and broadly neutralizing antibodies, (2) exploring the mechanisms of viral polyprotein processing, and (3) examining how the innate immune system distinguishes self versus viral RNAs. Our long-term goal is to discover and develop an effective HCV vaccine, novel antiviral drugs, and RIG-I immunomodulators for use as broad-based antiviral agents.

HCV represents an important public health problem, infecting an estimated 2% of the world’s population, approximately 150 million people. There is no vaccine against HCV and it is estimated that an additional 3-4 million individuals become newly infected each year worldwide. Several direct acting antivirals for HCV have recently been FDA approved; however, there is still a need for an HCV vaccine as these drugs are prohibitively expensive. HCV is an enveloped virus with an outer shell composed of many copies of two glycoproteins, E1 and E2. Mechanistic studies aimed at providing a detailed understanding of HCV entry have lagged behind due to an inability to produce properly folded HCV glycoproteins. To circumvent this obstacle, my laboratory has developed a cost-, labor-, and time-efficient method for the large-scale production of recombinant glycoproteins in mammalian cell lines with native, post-translational modifications. This production system enabled the first determination of the core domain of HCV E2 structure in complex with an Fab (Khan et al. Nature 2014). This structure revealed that HCV E2 does not share any similarity to other viral glycoproteins, including those from closely related viruses, suggesting that HCV may use a novel entry mechanism.

Numerous viruses, many of which severely impact human health around the globe (e.g. HIV, Zika virus, Dengue virus, West Nile virus, chikungunya virus, and SARS virus), utilize a gene expression mechanism wherein one gene encodes a single polyprotein that is post-translationally cleaved into individual proteins. Proteolytic processing occurs in a highly regulated and coordinated manner with intermediates having distinct functions and playing important roles during viral infection. Remarkably, viral proteases found within the polyprotein are capable of performing multiple cleavages that can be separated by long distances in linear sequence. Despite the commonality of polyprotein processing as a means of regulating the viral life cycle, very little is known about the higher order complexes of replication proteins post-processing, and even less about the structural properties of the precursor forms. Much of the existing biochemical and biophysical information on viral proteins stems from structural results of individual domains or mature forms of the viral proteins. To fully understand the biology and the regulation behind each step of the viral life cycle, it is absolutely necessary to examine the changes in the properties of viral proteins before and after polyprotein processing. We have determined the structure of the precleavage form of a portion of the Alphavirus replication machinery, helping to illuminate these critical steps in the virus life cycle (Shin et al. PNAS 2012). Our results have provided unprecedented insights into viral polyprotein processing and pathogenesis, which may be applicable to other important human viruses that undergo polyprotein processing.

The innate immune system is an evolutionarily ancient component of the host defense mechanisms. The innate immune receptors can efficiently distinguish general features of pathogens in a background of related self-molecules. The RIG-I (retinoic-acid-inducible gene-I) like receptors (RLR) consist of three cytoplasmic proteins (RIG-I, MDA5 and LGP2) that detect the presence of viral RNA in infected cells. A major goal of our research is to understand the mechanism of self versus non-self recognition and immune signaling actions of the RLR family receptors. To understand the synergy between the various domains of RIG-I for RNA binding and activation, my laboratory determined the structure of the RNA binding domains of human RIG-I in complex with dsRNA and an ATP analogue (Jiang et al. Nature, 2011). Recently, we established that the 2’O-methyl group on the first nucleotide adjacent to the 7-methyl guanosine cap is an important self versus non-self marker for RIG-I and demonstrated that RNAs with only 7-methyl guanosine caps are PAMPs (Devarkar et al. PNAS 2016).

Image showing Hepatitis C virus envelope glycoprotein 2 interacting with cellular receptor CD81

Hepatitis C virus envelope glycoprotein 2 interacting with cellular receptor CD81

Credit: NIAID
Selected Publications

Kumar A, Hossain RA, Yost SA, Bu W, Wang Y, Dearborn AD, Grakoui A, Cohen JI, Marcotrigiano J. Structural insights into hepatitis C virus receptor binding and entry. Nature. 2021 Oct;598(7881):521-525.

Devarkar SC, Wang C, Miller MT, Ramanathan A, Jiang F, Khan AG, Patel SS, Marcotrigiano J. Structural basis for m7G recognition and 2'-O-methyl discrimination in capped RNAs by the innate immune receptor RIG-I. Proc Natl Acad Sci U S A. 2016 Jan 19;113(3):596-601.

Khan AG, Whidby J, Miller MT, Scarborough H, Zatorski AV, Cygan A, Price AA, Yost SA, Bohannon CD, Jacob J, Grakoui A, Marcotrigiano J. Structure of the core ectodomain of the hepatitis C virus envelope glycoprotein 2. Nature. 2014 May 15;509(7500):381-4.

Shin G, Yost SA, Miller MT, Elrod EJ, Grakoui A, Marcotrigiano J. Structural and functional insights into alphavirus polyprotein processing and pathogenesis. Proc Natl Acad Sci U S A. 2012 Oct 9;109(41):16534-9.

Jiang F, Ramanathan A, Miller MT, Tang GQ, Gale M Jr, Patel SS, Marcotrigiano J. Structural basis of RNA recognition and activation by innate immune receptor RIG-I. Nature. 2011 Sep 25;479(7373):423-7.

Whidby J, Mateu G, Scarborough H, Demeler B, Grakoui A, Marcotrigiano J. Blocking hepatitis C virus infection with recombinant form of envelope protein 2 ectodomain. J Virol. 2009 Nov;83(21):11078-89.

Visit PubMed for a complete publication listing.

Major Areas of Research
  • Explore the mechanism of entry and replication of RNA viruses
  • Understand how the cell distinguishes self from non-self 
  • Characterize the immune response to RNA virus infection
  • Contribute to the development of novel therapies to combat infection and spread of RNA viruses
  • Develop novel methods for the recombinant production of challenging proteins in mammalian cells. 
Research Group Page

Hepatitis Disease-Specific Research

Hepatitis is an inflammation of the liver. Viruses are the most common cause of hepatitis, but the condition can also be caused by other infections, heavy alcohol use, toxins, certain medications, and autoimmune disease. There are five main virus types that cause hepatitis---type A, B, C, D, and E. Hepatitis A and E are typically caused by ingesting contaminated food or water. Hepatitis B commonly occurs through contact with infected blood, semen or other bodily fluid through sex, sharing needles or other drug-injection equipment or from mother to baby at birth. Hepatitis C is a blood-borne virus that is largely spread by sharing needles or other drug injection equipment. Hepatitis D, which is transmitted through contact with infectious blood, occurs only among people with hepatitis B infection.

Hepatitis B, C, and D afflict more than half a billion people worldwide and are responsible for more than a million deaths a year. Chronic infection with these viruses can lead to cirrhosis of the liver, end-stage liver disease, and liver cancer.

Hepatitis Research

NIAID supports and conducts research on each of the five known hepatitis viruses—A, B, C, D, and E. During the past 60 years, NIAID-supported investigators have been involved in many important advances in hepatitis research, including:

  • Discovery of the hepatitis A and E viruses
  • Development of one of the first diagnostic tests for hepatitis A
  • Studies that led to the creation of the hepatitis A vaccine
  • Studies that laid the foundation for advanced development of a hepatitis E vaccine.

NIAID’s research program emphasizes the study of hepatitis B and C viruses due to the large magnitude of medical burdens that they impose. Studies focus on understanding the immune response to infection, the course of disease development, and developing new therapeutics and vaccines for these viruses.

Related Public Health and Government Information

To learn about risk factors for hepatitis and current prevention and treatment strategies visit the MedlinePlus hepatitis site.

Photo of liver tissue specimen extracted from a viral hepatitis patient.
Hepatitis
Page Summary
Hepatitis is an inflammation of the liver. Viruses are the most common cause of hepatitis, but the condition can also be caused by other infections, heavy alcohol use, toxins, certain medications, and autoimmune disease. There are five main virus types that cause hepatitis---type A, B, C, D, and E.
Research Area Type
Diseases & Conditions

Patrizia Farci, M.D.

Chief, Hepatic Pathogenesis Section

Major Areas of Research

  • Pathogenesis of acute and chronic viral hepatitis
  • Molecular mechanisms of liver fibrosis progression and regression
  • Role of liver cirrhosis in the pathogenesis of hepatocellular carcinoma
  • Role of neutralizing antibodies in the prevention and control of hepatitis C virus (HCV) infection
  • HCV evolution and clinical outcome
  • Search for new hepatitis agents

Program Description

The Hepatic Pathogenesis Section (HPS) was established in 2010 to conduct translational research in the field of liver diseases, particularly in the study of pathogenesis of acute and chronic viral hepatitis, with the main goal of merging clinical medicine with basic research. Acute and chronic inflammatory liver diseases, especially viral hepatitis, are a major cause of morbidity and mortality both in the United States and in the rest of the world. A significant proportion of patients die of long-term sequelae of chronic liver disease, primarily cirrhosis and liver cancer.

Although there has been considerable progress in the control and treatment of acute and chronic viral hepatitis, future advances will depend on a more thorough knowledge of the mechanisms of pathogenesis. HPS has developed a comprehensive research program to study the molecular mechanisms of pathogenesis of acute liver failure and acute and chronic hepatitis especially of viral etiology, as well as the long-term sequelae of chronic hepatitis, cirrhosis, and hepatocellular carcinoma. Current research efforts also include the search for new hepatitis agents that may be implicated in the etiology of other forms of acute and chronic inflammatory liver diseases. New knowledge gained from these studies will pave the way toward the development of better means of preventing, diagnosing, and treating these illnesses.

Biography

Dr. Farci earned her M.D. at the University of Cagliari Medical School, Italy, and then became a board-certified specialist in infectious diseases and gastroenterology at the same university. She was trained at the department of gastroenterology of the Molinette Hospital in Torino under Dr. Mario Rizzetto and at the department of medicine of the Royal Free Hospital School of Medicine in London under Professor Sheila Sherlock. In 1989, she joined the laboratory of Dr. Robert H. Purcell in the Laboratory of Infectious Diseases (LID) as a visiting scientist. In 1992, she became associate professor of medicine and, in 2000, full professor of medicine and director of the liver unit and of the postgraduate school of gastroenterology at the University of Cagliari. In 2007, she returned to LID, where in 2010 she became chief of the Hepatic Pathogenesis Section.

Research Group

Farah Alayli, Zhaochun Chen, Emily J Danoff, Teresa Pollicino, Anna Pomerenke

Selected Publications

Melis M, Diaz G,  Kleiner DE, Zamboni F, Kabat J, Lai J, Mogavero G, Tice A, Engle RE, Becker S, Brown CR, Hanson JC, Rodrigues-Canales J, Emmert-Buck M, Govindarajan S, Kew M, Farci P. Viral expression and molecular profiling in liver tissue versus microdissected hepatocytes in hepatitis B virus-associated hepatocellular carcinoma. J Transl Med. 2014 Aug 21;12:230.

Diaz G, Melis M, Tice A, Kleiner DE, Mishra L, Zamboni F, Farci P. Identification of microRNAs specifically expressed in hepatitis C virus-associated hepatocellular carcinoma. Int J Cancer. 2013 Aug 15;133(4):816-24.

Farci P, Wollenberg K, Diaz G, Engle RE, Lai ME, Klenerman P, Purcell RH, Pybus OG, Alter HJ. Profibrogenic chemokines and viral evolution predict rapid progression of hepatitis C to cirrhosis. Proc Natl Acad Sci U S A. 2012 Sep 4;109(36):14562-7.

Farci P, Diaz G, Chen Z, Govindarajan S, Tice A, Agulto L, Pittaluga S, Boon D, Yu C, Engle RE, Haas M, Simon R, Purcell RH, Zamboni F. B-cell gene signature with massive intrahepatic production of antibodies to hepatitis B core antigen in hepatitis B virus-associated acute liver failureProc Natl Acad Sci U S A. 2010 May 11;107(19):8766-71.

Farci P, Quinti I, Farci S, Alter HJ, Strazzera R, Palomba E, Coiana A, Cao D, Casadei AM, Ledda R, Iorio R, Vegnente A, Diaz G, Tovo PA. Evolution of hepatitis C viral quasispecies and hepatic injury in perinatally infected children followed prospectivelyProc Natl Acad Sci U S A. 2006 May 30;103(22):8475-80.

Farci P, Shimoda A, Coiana A, Diaz G, Peddis G, Melpolder JC, Strazzera A, Chien DY, Munoz SJ, Balestrieri A, Purcell RH, Alter HJ. The outcome of acute hepatitis C predicted by the evolution of the viral quasispeciesScience. 2000 Apr 14;288(5464):339-44.

Visit PubMed for a complete publication listing.

Section or Unit Name
Hepatic Pathogenesis Section
First Name
Patrizia
Last Name
Farci
Suffix
M.D.
Exclude from directory
Off
This Researcher/Clinician’s Person Page
Program Description

The Hepatic Pathogenesis Section (HPS) was established in 2010 to conduct translational research in the field of liver diseases, particularly in the study of pathogenesis of acute and chronic viral hepatitis, with the main goal of merging clinical medicine with basic research. Acute and chronic inflammatory liver diseases, especially viral hepatitis, are a major cause of morbidity and mortality both in the United States and in the rest of the world. A significant proportion of patients die of long-term sequelae of chronic liver disease, primarily cirrhosis and liver cancer.

Although there has been considerable progress in the control and treatment of acute and chronic viral hepatitis, future advances will depend on a more thorough knowledge of the mechanisms of pathogenesis. HPS has developed a comprehensive research program to study the molecular mechanisms of pathogenesis of acute liver failure and acute and chronic hepatitis especially of viral etiology, as well as the long-term sequelae of chronic hepatitis, cirrhosis, and hepatocellular carcinoma. Current research efforts also include the search for new hepatitis agents that may be implicated in the etiology of other forms of acute and chronic inflammatory liver diseases. New knowledge gained from these studies will pave the way toward the development of better means of preventing, diagnosing, and treating these illnesses.

Selected Publications

Melis M, Diaz G,  Kleiner DE, Zamboni F, Kabat J, Lai J, Mogavero G, Tice A, Engle RE, Becker S, Brown CR, Hanson JC, Rodrigues-Canales J, Emmert-Buck M, Govindarajan S, Kew M, Farci P. Viral expression and molecular profiling in liver tissue versus microdissected hepatocytes in hepatitis B virus-associated hepatocellular carcinoma. J Transl Med. 2014 Aug 21;12:230.

Diaz G, Melis M, Tice A, Kleiner DE, Mishra L, Zamboni F, Farci P. Identification of microRNAs specifically expressed in hepatitis C virus-associated hepatocellular carcinoma. Int J Cancer. 2013 Aug 15;133(4):816-24.

Farci P, Wollenberg K, Diaz G, Engle RE, Lai ME, Klenerman P, Purcell RH, Pybus OG, Alter HJ. Profibrogenic chemokines and viral evolution predict rapid progression of hepatitis C to cirrhosis. Proc Natl Acad Sci U S A. 2012 Sep 4;109(36):14562-7.

Farci P, Diaz G, Chen Z, Govindarajan S, Tice A, Agulto L, Pittaluga S, Boon D, Yu C, Engle RE, Haas M, Simon R, Purcell RH, Zamboni F. B-cell gene signature with massive intrahepatic production of antibodies to hepatitis B core antigen in hepatitis B virus-associated acute liver failureProc Natl Acad Sci U S A. 2010 May 11;107(19):8766-71.

Farci P, Quinti I, Farci S, Alter HJ, Strazzera R, Palomba E, Coiana A, Cao D, Casadei AM, Ledda R, Iorio R, Vegnente A, Diaz G, Tovo PA. Evolution of hepatitis C viral quasispecies and hepatic injury in perinatally infected children followed prospectivelyProc Natl Acad Sci U S A. 2006 May 30;103(22):8475-80.

Farci P, Shimoda A, Coiana A, Diaz G, Peddis G, Melpolder JC, Strazzera A, Chien DY, Munoz SJ, Balestrieri A, Purcell RH, Alter HJ. The outcome of acute hepatitis C predicted by the evolution of the viral quasispeciesScience. 2000 Apr 14;288(5464):339-44.

Visit PubMed for a complete publication listing.

Additional Information

Research Group

Farah Alayli

Zhaochun Chen

Emily J Danoff

Teresa Pollicino

Anna Pomerenke

Research Group Description

Farah Alayli, Zhaochun Chen, Emily J Danoff, Teresa Pollicino, Anna Pomerenke

Major Areas of Research
  • Pathogenesis of acute and chronic viral hepatitis
  • Molecular mechanisms of liver fibrosis progression and regression
  • Role of liver cirrhosis in the pathogenesis of hepatocellular carcinoma
  • Role of neutralizing antibodies in the prevention and control of hepatitis C virus (HCV) infection
  • HCV evolution and clinical outcome
  • Search for new hepatitis agents