The effort to investigate the gender differences in lupus has been mostly devoted to the cellular and humoral immune responses and much less to the cellular damage. Recently cell death, the ultimate cell damage, has been recognized as a fundamental pathogenic mechanism in lupus glomerulonephritis (GN). GN is among the most devastating effects of lupus disease. It is the leading cause of long-term disability, and ranks high as a cause of morbidity and mortality. Despite the striking 9:1 female to male lupus ratio, GN is more severe in males, suggesting that the molecular pathways in both sexes involved in the renal damage must be as aggressive. We have recently discovered that the inhibition or deletion of poly (ADP) ribose polymerase (PARP) 1, an enzyme involved in necrotic cell death and production of pro-inflammatory cytokines, protects only male mice from lupus GN, demonstrating the principle that males and females employ different pathways of cellular damage downstream the immune complex deposition, a fundamental initiator of renal damage. Therefore the objective of this application is to investigate the molecular pathways that induce tissue damage, necrotic cell death and inflammation in males and females during lupus GN. We propose that cell death during lupus GN is a fundamental renal intrinsic pathogenic mechanism that actively participates to the disease progression. We also propose that during GN there is a balance between the pro-inflammatory necrotic death and the anti-inflammatory apoptosis, and when this balance is toward necrosis the disease is more severe and progresses at a faster pace.
In AIMs I and II we intend to demonstrate in mouse models of lupus GN that these pathways are regulated by two major factors: 1) PARP-1, which induces necrosis, increases the release of pro-inflammatory cytokines, such as high molecular group box (HMGB)-1, modulates the expression of adhesion molecules, and facilitates the activation of NF?B in situ~ and 2) estrogens, which modulate the expression of adhesion molecules, cytokines and the induction of apoptotic versus necrotic cell death via inactivation of PARP-1. Because there are two major pathways that lead to necrotic cell death, PARP-1-dependent and Receptor Interacting Protein (RIP)-1 and -3-dependent, and because necrosis also occurs in females, in AIM I and II we also intend to determine if females have proclivity for RIP-1/3-dependent necrosis. In the third AIM we will test the relevance of necrotic cell death pathways in human lupus nephritis. We will test the hypothesis that HMGB1 released during nephritis might be a biomarker in human lupus GN. The rationale for our project is that a better understanding of the pathways regulating the tissue damage in lupus GN pathophysiologies in males and females will lead to a tailored and better treatment for each gender.

Public Health Relevance

Lupus has its most devastating complication in the kidney, which is a leading cause of morbidity and mortality. One of the most important characteristics of kidney damage in lupus is the presence of necrotic lesions. We intend to demonstrate that sex and sexual hormones play a fundamental role in controlling the necrotic lesions and that a better understanding of this process will provide the rational for a therapy targeting necrosis.

Agency
National Institute of Health (NIH)
Institute
National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS)
Type
Research Project (R01)
Project #
5R01AR061569-03
Application #
8702085
Study Section
Hypersensitivity, Autoimmune, and Immune-mediated Diseases Study Section (HAI)
Program Officer
Mancini, Marie
Project Start
2012-08-01
Project End
2017-07-31
Budget Start
2014-08-01
Budget End
2015-07-31
Support Year
3
Fiscal Year
2014
Total Cost
Indirect Cost
Name
Temple University
Department
Internal Medicine/Medicine
Type
Schools of Medicine
DUNS #
City
Philadelphia
State
PA
Country
United States
Zip Code
19122
Corradetti, Chelsea; Jog, Neelakshi R; Cesaroni, Matteo et al. (2018) Estrogen Receptor ? Signaling Exacerbates Immune-Mediated Nephropathies through Alteration of Metabolic Activity. J Immunol 200:512-522
Jog, N R; Blanco, I; Lee, I et al. (2016) Urinary high-mobility group box-1 associates specifically with lupus nephritis class V. Lupus 25:1551-1557
Meng, Lijun; Hu, Shaoyan; Wang, Jian et al. (2016) DLL4+ dendritic cells: Key regulators of Notch Signaling in effector T cell responses. Pharmacol Res 113:449-457
Corradetti, Chelsea; Jog, Neelakshi R; Gallucci, Stefania et al. (2016) Immune-Mediated Nephropathy and Systemic Autoimmunity in Mice Does Not Require Receptor Interacting Protein Kinase 3 (RIPK3). PLoS One 11:e0163611
Meng, Lijun; Bai, Zhenjiang; He, Shan et al. (2016) The Notch Ligand DLL4 Defines a Capability of Human Dendritic Cells in Regulating Th1 and Th17 Differentiation. J Immunol 196:1070-80
Gallo, Paul M; Rapsinski, Glenn J; Wilson, R Paul et al. (2015) Amyloid-DNA Composites of Bacterial Biofilms Stimulate Autoimmunity. Immunity 42:1171-84
Shanmughapriya, Santhanam; Rajan, Sudarsan; Hoffman, Nicholas E et al. (2015) Ca2+ signals regulate mitochondrial metabolism by stimulating CREB-mediated expression of the mitochondrial Ca2+ uniporter gene MCU. Sci Signal 8:ra23
Jog, Neelakshi R; Caricchio, Roberto (2015) 17? estradiol regulates adhesion molecule expression in mesangial cells during glomerulonephritis. Clin Immunol 159:13-22
Jog, Neelakshi R; Caricchio, Roberto (2014) The role of necrotic cell death in the pathogenesis of immune mediated nephropathies. Clin Immunol 153:243-53
Jog, N R; Caricchio, R (2013) Differential regulation of cell death programs in males and females by Poly (ADP-Ribose) Polymerase-1 and 17? estradiol. Cell Death Dis 4:e758

Showing the most recent 10 out of 12 publications