1
|
Hickey MJ, Sudhakar V. Looking below the surface: using intravital imaging to decipher inflammatory renal disease and renal cell injury. Am J Physiol Renal Physiol 2025; 328:F418-F430. [PMID: 39918796 DOI: 10.1152/ajprenal.00321.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 11/22/2024] [Accepted: 02/02/2025] [Indexed: 03/04/2025] Open
Abstract
Renal function can be perturbed by a range of stimuli that cause cellular injury and inflammation in the kidney. These injurious and inflammatory processes are typically dynamic and progressive, involving the actions of highly migratory cells such as leukocytes and cellular responses that occur over time spans ranging from seconds to weeks. Understanding these dynamic responses has entailed the use of imaging technologies that allow visualization and capture of events over different time spans, ideally in intact organs in live, experimental animals. The technique that allows this is intravital imaging. Intravital imaging, particularly multiphoton intravital microscopy, has been crucial to the investigation of dynamic physiological and pathophysiological processes in the kidney for many years, driving key developments in our understanding of renal (patho)physiology. This includes the mechanisms of ultrafiltrate generation, the response to acute kidney injury, and how inflammatory leukocytes are recruited to and cause injury in the kidney. This review describes the key studies that have applied intravital imaging to the investigation of models of inflammatory renal disease. The responses examined include those restricted to the glomerulus and the effects of acute kidney injury on the tubulointerstitium. Future innovations and directions in this field of research are also discussed.
Collapse
Affiliation(s)
- Michael J Hickey
- Centre for Inflammatory Diseases, Department of Medicine, Monash Medical Centre, Monash University, Clayton, Victoria, Australia
| | - Vaishnavi Sudhakar
- Centre for Inflammatory Diseases, Department of Medicine, Monash Medical Centre, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
2
|
Huang F, Lv Y, Liu S, Wu H, Liu Q. Animal models for anti-neutrophil cytoplasmic antibody-associated vasculitis: Are current models good enough? Animal Model Exp Med 2023; 6:452-463. [PMID: 37614099 PMCID: PMC10614129 DOI: 10.1002/ame2.12345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 07/22/2023] [Indexed: 08/25/2023] Open
Abstract
Antineutrophil cytoplasmic autoantibody (ANCA)-associated vasculitis (AAV) is a rare and severe systemic autoimmune disease characterized by pauci-immune necrotizing inflammation of small blood vessels. AAV involves multiple organ systems throughout the body. Our knowledge of the pathogenesis of AAV has increased considerably in recent years, involving cellular, molecular and genetic factors. Because of the controlled environment with no other confounding factors, animal models are beneficial for studying the mechanistic details of disease development and for providing novel therapeutic targets with fewer toxic side effects. However, the complexity and heterogeneity of AAV make it very difficult to establish a single animal model that can fully represent the entire clinical spectrum found in patients. The aim of this review is to overview the current status of animal models for AAV, outline the pros and cons of methods, and propose potential directions for future research.
Collapse
Affiliation(s)
- Fei Huang
- Department of GeriatricsTongji Hospital, Tongji Medical college, Huazhong University of Science and TechnologyWuhanChina
- Department of General MedicineTongji Hospital, Tongji Medical college, Huazhong University of Science and TechnologyWuhanChina
| | - Yongman Lv
- Department of NephrologyTongji Hospital, Tongji Medical college, Huazhong University of Science and TechnologyWuhanChina
- Department of health management centerTongji Hospital, Tongji Medical college, Huazhong University of Science and TechnologyWuhanChina
| | - Siyang Liu
- Department of NephrologyTongji Hospital, Tongji Medical college, Huazhong University of Science and TechnologyWuhanChina
| | - Hao Wu
- Interdisciplinary Sciences InstituteHuazhong Agricultural UniversityWuhanChina
- State Key Laboratory of Agricultural Microbiology, College of Veterinary MedicineHuazhong Agricultural UniversityWuhanChina
| | - Qingquan Liu
- Department of NephrologyTongji Hospital, Tongji Medical college, Huazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
3
|
Shioda R, Jo-Watanabe A, Okuno T, Saeki K, Nakayama M, Suzuki Y, Yokomizo T. The leukotriene B 4 /BLT1-dependent neutrophil accumulation exacerbates immune complex-mediated glomerulonephritis. FASEB J 2023; 37:e22789. [PMID: 36692419 DOI: 10.1096/fj.202201936r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 12/25/2022] [Accepted: 01/11/2023] [Indexed: 01/25/2023]
Abstract
Crescent formation is the most important pathological finding that defines the prognosis of nephritis. Although neutrophils are known to play an important role in the progression of crescentic glomerulonephritis, such as anti-neutrophil cytoplasmic antibody (ANCA)-associated glomerulonephritis, the key chemoattractant for neutrophils in ANCA-associated glomerulonephritis has not been identified. Here, we demonstrate that a lipid chemoattractant, leukotriene B4 (LTB4 ), and its receptor BLT1 are primarily involved in disease pathogenesis in a mouse model of immune complex-mediated crescentic glomerulonephritis. Circulating neutrophils accumulated into glomeruli within 1 h after disease onset, which was accompanied by LTB4 accumulation in the kidney cortex, leading to kidney injury. LTB4 was produced by cross-linking of Fc gamma receptors on neutrophils. Mice deficient in BLT1 or LTB4 biosynthesis exhibited suppressed initial neutrophil infiltration and subsequent thrombotic glomerulonephritis and renal fibrosis. Depletion of neutrophils before, but not after, disease onset prevented proteinuria and kidney injury, indicating the essential role of neutrophils in the early phase of glomerulonephritis. Administration of a BLT1 antagonist before and after disease onset almost completely suppressed induction of glomerulonephritis. Finally, we found that the glomeruli from patients with ANCA-associated glomerulonephritis contained more BLT1-positive cells than glomeruli from patients with other etiologies. Taken together, the LTB4 -BLT1 axis is the key driver of neutrophilic glomerular inflammation, and will be a novel therapeutic target for the crescentic glomerulonephritis.
Collapse
Affiliation(s)
- Ryotaro Shioda
- Department of Biochemistry, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Department of Nephrology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Airi Jo-Watanabe
- Department of Biochemistry, Juntendo University Graduate School of Medicine, Tokyo, Japan.,AMED-PRIME, Japan Agency for Medical Research and Development, Tokyo, Japan
| | - Toshiaki Okuno
- Department of Biochemistry, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Kazuko Saeki
- Department of Biochemistry, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Maiko Nakayama
- Department of Nephrology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Yusuke Suzuki
- Department of Nephrology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Takehiko Yokomizo
- Department of Biochemistry, Juntendo University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
4
|
Saito S, Tatsumoto N, Cao DY, Nosaka N, Nishi H, Leal DN, Bernstein E, Shimada K, Arditi M, Bernstein KE, Yamashita M. Overexpressed angiotensin-converting enzyme in neutrophils suppresses glomerular damage in crescentic glomerulonephritis. Am J Physiol Renal Physiol 2022; 323:F411-F424. [PMID: 35979968 PMCID: PMC9484997 DOI: 10.1152/ajprenal.00067.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 07/22/2022] [Accepted: 08/13/2022] [Indexed: 11/22/2022] Open
Abstract
While angiotensin-converting enzyme (ACE) regulates blood pressure by producing angiotensin II as part of the renin-angiotensin system, we recently reported that elevated ACE in neutrophils promotes an effective immune response and increases resistance to infection. Here, we investigate if such neutrophils protect against renal injury in immune complex (IC)-mediated crescentic glomerulonephritis (GN) through complement. Nephrotoxic serum nephritis (NTN) was induced in wild-type and NeuACE mice that overexpress ACE in neutrophils. Glomerular injury of NTN in NeuACE mice was attenuated with much less proteinuria, milder histological injury, and reduced IC deposits, but presented with more glomerular neutrophils in the early stage of the disease. There were no significant defects in T and B cell functions in NeuACE mice. NeuACE neutrophils exhibited enhanced IC uptake with elevated surface expression of FcγRII/III and complement receptor CR1/2. IC uptake in neutrophils was enhanced by NeuACE serum containing elevated complement C3b. Given no significant complement activation by ACE, this suggests that neutrophil ACE indirectly preactivates C3 and that the C3b-CR1/2 axis and elevated FcγRII/III play a central role in IC elimination by neutrophils, resulting in reduced glomerular injury. The present study identified a novel renoprotective role of ACE in glomerulonephritis; elevated neutrophilic ACE promotes elimination of locally formed ICs in glomeruli via C3b-CR1/2 and FcγRII/III, ameliorating glomerular injury.NEW & NOTEWORTHY We studied immune complex (IC)-mediated crescentic glomerulonephritis in NeuACE mice that overexpress ACE only in neutrophils. Such mice show no significant defects in humoral immunity but strongly resist nephrotoxic serum nephritis (less proteinuria, milder histological damage, reduced IC deposits, and more glomerular neutrophils). NeuACE neutrophils enhanced IC uptake via increased surface expression of CR1/2 and FcgRII/III, as well as elevated serum complement C3b. These results suggest neutrophil ACE as a novel approach to reducing glomerulonephritis.
Collapse
Affiliation(s)
- Suguru Saito
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California
| | - Narihito Tatsumoto
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Duo-Yao Cao
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California
| | - Nobuyuki Nosaka
- Division of Infectious Diseases and Immunology, Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, California
| | - Hiroshi Nishi
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Daniel N Leal
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Ellen Bernstein
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California
| | - Kenichi Shimada
- Division of Infectious Diseases and Immunology, Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, California
- Department of Biomedical Sciences, Infectious and Immunologic Diseases Research Center, Cedars-Sinai Medical Center, Los Angeles, California
| | - Moshe Arditi
- Division of Infectious Diseases and Immunology, Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, California
- Department of Biomedical Sciences, Infectious and Immunologic Diseases Research Center, Cedars-Sinai Medical Center, Los Angeles, California
| | - Kenneth E Bernstein
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, California
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California
| | - Michifumi Yamashita
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| |
Collapse
|
5
|
Melica ME, Antonelli G, Semeraro R, Angelotti ML, Lugli G, Landini S, Ravaglia F, La Regina G, Conte C, De Chiara L, Peired AJ, Mazzinghi B, Donati M, Molli A, Steiger S, Magi A, Bartalucci N, Raglianti V, Guzzi F, Maggi L, Annunziato F, Burger A, Lazzeri E, Anders HJ, Lasagni L, Romagnani P. Differentiation of crescent-forming kidney progenitor cells into podocytes attenuates severe glomerulonephritis in mice. Sci Transl Med 2022; 14:eabg3277. [PMID: 35947676 PMCID: PMC7614034 DOI: 10.1126/scitranslmed.abg3277] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Crescentic glomerulonephritis is characterized by vascular necrosis and parietal epithelial cell hyperplasia in the space surrounding the glomerulus, resulting in the formation of crescents. Little is known about the molecular mechanisms driving this process. Inducing crescentic glomerulonephritis in two Pax2Cre reporter mouse models revealed that crescents derive from clonal expansion of single immature parietal epithelial cells. Preemptive and delayed histone deacetylase inhibition with panobinostat, a drug used to treat hematopoietic stem cell disorders, attenuated crescentic glomerulonephritis with recovery of kidney function in the two mouse models. Three-dimensional confocal microscopy and stimulated emission depletion superresolution imaging of mouse glomeruli showed that, in addition to exerting an anti-inflammatory and immunosuppressive effect, panobinostat induced differentiation of an immature hyperplastic parietal epithelial cell subset into podocytes, thereby restoring the glomerular filtration barrier. Single-cell RNA sequencing of human renal progenitor cells in vitro identified an immature stratifin-positive cell subset and revealed that expansion of this stratifin-expressing progenitor cell subset was associated with a poor outcome in human crescentic glomerulonephritis. Treatment of human parietal epithelial cells in vitro with panobinostat attenuated stratifin expression in renal progenitor cells, reduced their proliferation, and promoted their differentiation into podocytes. These results offer mechanistic insights into the formation of glomerular crescents and demonstrate that selective targeting of renal progenitor cells can attenuate crescent formation and the deterioration of kidney function in crescentic glomerulonephritis in mice.
Collapse
Affiliation(s)
- Maria Elena Melica
- Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), University of Florence, Florence 50139, Italy,Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy
| | - Giulia Antonelli
- Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), University of Florence, Florence 50139, Italy,Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy
| | - Roberto Semeraro
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Maria Lucia Angelotti
- Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), University of Florence, Florence 50139, Italy,Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy
| | - Gianmarco Lugli
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy,Nephrology and Dialysis Unit, Meyer Children’s Hospital, Florence 50139, Italy
| | - Samuela Landini
- Nephrology and Dialysis Unit, Meyer Children’s Hospital, Florence 50139, Italy
| | - Fiammetta Ravaglia
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy
| | - Gilda La Regina
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy
| | - Carolina Conte
- Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), University of Florence, Florence 50139, Italy,Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy
| | - Letizia De Chiara
- Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), University of Florence, Florence 50139, Italy,Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy
| | - Anna Julie Peired
- Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), University of Florence, Florence 50139, Italy,Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy
| | - Benedetta Mazzinghi
- Nephrology and Dialysis Unit, Meyer Children’s Hospital, Florence 50139, Italy
| | - Marta Donati
- Nephrology and Dialysis Unit, Meyer Children’s Hospital, Florence 50139, Italy
| | - Alice Molli
- Nephrology and Dialysis Unit, Meyer Children’s Hospital, Florence 50139, Italy
| | - Stefanie Steiger
- Division of Nephrology, Medizinische Klinik and Poliklinik IV, Klinikum der LMU München, Munich 80336, Germany
| | - Alberto Magi
- Department of Information Engineering, University of Florence, Florence, Italy
| | - Niccolò Bartalucci
- Department of Experimental and Clinical Medicine, CRIMM, Center Research and Innovation of Myeloproliferative Neoplasms, AOUC, University of Florence, Florence 50139, Italy
| | - Valentina Raglianti
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy,Nephrology and Dialysis Unit, Meyer Children’s Hospital, Florence 50139, Italy
| | - Francesco Guzzi
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy,Nephrology and Dialysis Unit, Meyer Children’s Hospital, Florence 50139, Italy
| | - Laura Maggi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Francesco Annunziato
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Alexa Burger
- Section of Developmental Biology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Elena Lazzeri
- Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), University of Florence, Florence 50139, Italy,Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy
| | - Hans-Joachim Anders
- Division of Nephrology, Medizinische Klinik and Poliklinik IV, Klinikum der LMU München, Munich 80336, Germany
| | - Laura Lasagni
- Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), University of Florence, Florence 50139, Italy,Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy,Corresponding authors. and
| | - Paola Romagnani
- Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), University of Florence, Florence 50139, Italy,Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy,Nephrology and Dialysis Unit, Meyer Children’s Hospital, Florence 50139, Italy,Corresponding authors. and
| |
Collapse
|
6
|
Animal models of vasculitis. Curr Opin Rheumatol 2022; 34:10-17. [PMID: 34783711 DOI: 10.1097/bor.0000000000000848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW Vasculitis describes a wide spectrum of rare, inflammatory, multisystem disorders. These heterogenous diseases all have inflammation of blood vessels as a central feature. However, they differ in terms of their genetic and environmental risk factors, disease pathogenesis, clinical presentations and treatment strategies. Many animal models of vasculitis exist, each resembling a different human clinical phenotype. This review provides an overview of recently published findings from experimental animal models of vasculitis. RECENT FINDINGS Several new animal models have been described during the review period. New insights gleaned from existing animal models regarding cause, disease effector mechanisms and novel treatments identified in established animal models are discussed. SUMMARY Animal models continue to be an important tool for understanding disease pathogenesis, especially in rare and complex diseases such as vasculitis. They also provide an invaluable platform for development and preclinical testing of new treatments.
Collapse
|
7
|
Al Mushafi A, Ooi JD, Odobasic D. Crescentic Glomerulonephritis: Pathogenesis and Therapeutic Potential of Human Amniotic Stem Cells. Front Physiol 2021; 12:724186. [PMID: 34721059 PMCID: PMC8554237 DOI: 10.3389/fphys.2021.724186] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Accepted: 09/24/2021] [Indexed: 12/15/2022] Open
Abstract
Chronic kidney disease (CKD) leads to significant morbidity and mortality worldwide. Glomerulonephritis (GN) is the second leading cause of CKD resulting in end stage renal failure. The most severe and rapidly progressive type of GN is characterized by glomerular crescent formation. The current therapies for crescentic GN, which consist of broad immunosuppressive drugs, are partially effective, non-specific, toxic and cause many serious side effects including infections, cancer, and cardiovascular problems. Therefore, new and safer therapies are needed. Human amniotic epithelial cells (hAECs) are a type of stem cell which are isolated from the placenta after birth. They represent an attractive and novel therapeutic option for the treatment of various inflammatory conditions owing to their unique and selective immunosuppressive ability, as well as their excellent safety profile and clinical applicability. In this review, we will discuss the immunopathogenesis of crescentic GN, issues with currently available treatments and how hAECs offer potential to become a new and harmless treatment option for this condition.
Collapse
Affiliation(s)
- Ahmed Al Mushafi
- Department of Medicine, Monash Medical Centre, Centre for Inflammatory Diseases, Monash University, Clayton, VIC, Australia
| | - Joshua D Ooi
- Department of Medicine, Monash Medical Centre, Centre for Inflammatory Diseases, Monash University, Clayton, VIC, Australia
| | - Dragana Odobasic
- Department of Medicine, Monash Medical Centre, Centre for Inflammatory Diseases, Monash University, Clayton, VIC, Australia
| |
Collapse
|
8
|
Sakai R, Ito M, Komai K, Iizuka-Koga M, Matsuo K, Nakayama T, Yoshie O, Amano K, Nishimasu H, Nureki O, Kubo M, Yoshimura A. Kidney GATA3 + regulatory T cells play roles in the convalescence stage after antibody-mediated renal injury. Cell Mol Immunol 2021; 18:1249-1261. [PMID: 32917984 PMCID: PMC8093306 DOI: 10.1038/s41423-020-00547-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 08/24/2020] [Indexed: 12/17/2022] Open
Abstract
FoxP3+ regulatory T cells (Tregs) play crucial roles in peripheral immune tolerance. In addition, Tregs that reside or accumulate in nonlymphoid tissues, called tissue Tregs, exhibit tissue-specific functions and contribute to the maintenance of tissue homeostasis and repair. In an experimental mouse model of crescentic glomerulonephritis induced by an anti-glomerular basement membrane antibody, Tregs started to accumulate in the kidney on day 10 of disease onset and remained at high levels (~30-35% of CD4+ T cells) during the late stage (days 21-90), which correlated with stable disease control. Treg depletion on day 21 resulted in the relapse of renal dysfunction and an increase in Th1 cells, suggesting that Tregs are essential for disease control during the convalescence stage. The Tregs that accumulated in the kidney showed tissue Treg phenotypes, including high expression of GATA3, ST2 (the IL33 receptor subunit), amphiregulin (Areg), and PPARγ. Although T-bet+ Tregs and RORγt+ Tregs were observed in the kidney, GATA3+ Tregs were predominant during the convalescence stage, and a PPARγ agonist enhanced the accumulation of GATA3+ Tregs in the kidney. To understand the function of specific genes in kidney Tregs, we developed a novel T cell transfer system to T cell-deficient mice. This experiment demonstrates that ST2, Areg, and CCR4 in Tregs play important roles in the accumulation of GATA3+ Tregs in the kidney and in the amelioration of renal injury. Our data suggest that GATA3 is important for the recruitment of Tregs into the kidney, which is necessary for convalescence after renal tissue destruction.
Collapse
Affiliation(s)
- Ryota Sakai
- Department of Microbiology and Immunology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.
- Department of Rheumatology and Clinical Immunology, Saitama Medical Center, Saitama Medical University, 1981 Kamoda, Kawagoe, 350-8550, Japan.
| | - Minako Ito
- Medical Institute of Bioregulation Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Kyoko Komai
- Department of Microbiology and Immunology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Mana Iizuka-Koga
- Department of Microbiology and Immunology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Kazuhiko Matsuo
- Division of Chemotherapy, Kindai University Faculty of Pharmacy, Higashi-Osaka, 577-8502, Japan
| | - Takashi Nakayama
- Division of Chemotherapy, Kindai University Faculty of Pharmacy, Higashi-Osaka, 577-8502, Japan
| | - Osamu Yoshie
- The Health and Kampo Institute, Sendai, Miyagi, 981-3205, Japan
| | - Koichi Amano
- Department of Rheumatology and Clinical Immunology, Saitama Medical Center, Saitama Medical University, 1981 Kamoda, Kawagoe, 350-8550, Japan
| | - Hiroshi Nishimasu
- Department of Biological Science, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Osamu Nureki
- Department of Biological Science, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Masato Kubo
- Center for Animal Disease Models, Research Institute for Biomedical Science, Tokyo University of Science, 2669 Yamazaki, Noda-shi, Chiba, 278-0022, Japan
| | - Akihiko Yoshimura
- Department of Microbiology and Immunology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.
| |
Collapse
|
9
|
Sakai R, Ito M, Yoshimoto K, Chikuma S, Kurasawa T, Kondo T, Suzuki K, Takeuchi T, Amano K, Yoshimura A. Tocilizumab monotherapy uncovered the role of the CCL22/17-CCR4 + Treg axis during remission of crescentic glomerulonephritis. Clin Transl Immunology 2020; 9:e1203. [PMID: 33163184 PMCID: PMC7596393 DOI: 10.1002/cti2.1203] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 09/08/2020] [Accepted: 10/08/2020] [Indexed: 12/11/2022] Open
Abstract
Objectives Tocilizumab (TCZ) is a humanised anti‐interleukin (IL)‐6 receptor (IL‐6R) monoclonal antibody that is a promising agent to treat various autoimmune diseases. However, the mechanism of TCZ efficacy is unclear. This study aims to elucidate the relationship between Tregs and IL‐6R blockade in autoimmunity‐mediated renal disease based on a TCZ‐treated cohort of patients with anti‐neutrophil cytoplasmic antibody (ANCA)‐associated vasculitis (AAV) and in an experimental model of crescentic glomerulonephritis (cGN). Methods We examined multiple serum levels of cytokines and chemokines and peripheral blood mononuclear cells in patients with AAV who received TCZ monotherapy and achieved drug‐free remission. Moreover, we investigated the mechanistic role of IL‐6R blockade in accelerated cGN model to analyse the local sites of inflammation. Results Serum chemokines CCL22 and CCL17, in addition to the CCR4+Foxp3+ Treg population, increased in patients who demonstrated drug‐free remission after the cessation of TCZ. In the cGN model, IL‐6R blockade ameliorated the disease, elevated CCL22/17 in CD206+CD11b+CD11c+ kidney M2‐like type macrophages, and increased the migration of Tregs into the kidney and regional lymph nodes. The local administration of CCL22 in the kidney facilitated Treg accumulation and reduced glomerular crescent formation. Conclusions This study revealed a new mechanism whereby effector Tregs migrate into the inflammatory kidney via the CCL22/17–CCR4 axis that is facilitated by M2‐like type macrophages that are induced by IL‐6R blockade.
Collapse
Affiliation(s)
- Ryota Sakai
- Department of Microbiology and Immunology Keio University School of Medicine Tokyo Japan.,Department of Rheumatology and Clinical Immunology Saitama Medical Center Saitama Medical University Kawagoe Japan
| | - Minako Ito
- Department of Microbiology and Immunology Keio University School of Medicine Tokyo Japan
| | - Keiko Yoshimoto
- Division of Rheumatology Department of Internal Medicine Keio University School of Medicine Tokyo Japan
| | - Shunsuke Chikuma
- Department of Microbiology and Immunology Keio University School of Medicine Tokyo Japan
| | - Takahiko Kurasawa
- Department of Rheumatology and Clinical Immunology Saitama Medical Center Saitama Medical University Kawagoe Japan
| | - Tsuneo Kondo
- Department of Rheumatology and Clinical Immunology Saitama Medical Center Saitama Medical University Kawagoe Japan
| | - Katsuya Suzuki
- Division of Rheumatology Department of Internal Medicine Keio University School of Medicine Tokyo Japan
| | - Tsutomu Takeuchi
- Division of Rheumatology Department of Internal Medicine Keio University School of Medicine Tokyo Japan
| | - Koichi Amano
- Department of Rheumatology and Clinical Immunology Saitama Medical Center Saitama Medical University Kawagoe Japan
| | - Akihiko Yoshimura
- Department of Microbiology and Immunology Keio University School of Medicine Tokyo Japan
| |
Collapse
|
10
|
Shochet L, Holdsworth S, Kitching AR. Animal Models of ANCA Associated Vasculitis. Front Immunol 2020; 11:525. [PMID: 32373109 PMCID: PMC7179669 DOI: 10.3389/fimmu.2020.00525] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 03/09/2020] [Indexed: 01/05/2023] Open
Abstract
Anti-neutrophil cytoplasmic antibody (ANCA) associated vasculitis (AAV) is a rare and severe autoimmune multisystemic disease. Its pathogenesis involves multiple arms of the immune system, as well as complex interactions between immune cells and target organs. Experimental animal models of disease can provide the crucial link from human disease to translational research into new therapies. This is particularly true in AAV, due to low disease incidence and substantial disease heterogeneity. Animal models allow for controlled environments in which disease mechanisms can be defined, without the clinical confounders of environmental and lifestyle factors. To date, multiple animal models have been developed, each of which shed light on different disease pathways. Results from animal studies of AAV have played a crucial role in enhancing our understanding of disease mechanisms, and have provided direction toward newer targeted therapies. This review will summarize our understanding of AAV pathogenesis as has been gleaned from currently available animal models, as well as address their strengths and limitations. We will also discuss the potential for current and new animal models to further our understanding of this important condition.
Collapse
Affiliation(s)
- Lani Shochet
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, VIC, Australia.,Department of Nephrology, Monash Health, Clayton, VIC, Australia
| | - Stephen Holdsworth
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, VIC, Australia.,Department of Nephrology, Monash Health, Clayton, VIC, Australia.,Department of Immunology, Monash Health, Clayton, VIC, Australia
| | - A Richard Kitching
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, VIC, Australia.,Department of Nephrology, Monash Health, Clayton, VIC, Australia.,Department of Pediatric Nephrology, Monash Health, Clayton, VIC, Australia
| |
Collapse
|
11
|
Kitching AR, Holdsworth SR. The renal draining lymph nodes in acute inflammatory kidney disease. Kidney Int 2019; 95:254-256. [PMID: 30665562 DOI: 10.1016/j.kint.2018.10.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 10/05/2018] [Indexed: 11/15/2022]
Abstract
Renal lymphatics are implicated in renal disease, but their function in inflammatory kidney diseases is relatively poorly defined. In the current issue, Kasinath et al. examine the role of the kidney draining lymph node in experimental glomerulonephritis, as well the role of fibroblastic reticular cells within lymph nodes. Removing the kidney-draining lymph nodes prior to the induction of glomerulonephritis attenuated disease, as did interventions that affected the function of lymph nodes in general.
Collapse
Affiliation(s)
- A Richard Kitching
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, Victoria, Australia; Department of Nephrology, Monash Health, Clayton, Victoria, Australia; Department of Pediatric Nephrology, Monash Health, Clayton, Victoria, Australia.
| | - Stephen R Holdsworth
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, Victoria, Australia; Department of Nephrology, Monash Health, Clayton, Victoria, Australia; Department of Clinical Immunology, Monash Health, Clayton, Victoria, Australia
| |
Collapse
|
12
|
Kitching AR, Alikhan MA. CD8+ cells and glomerular crescent formation: outside-in as well as inside-out. J Clin Invest 2018; 128:3231-3233. [PMID: 29985169 DOI: 10.1172/jci122045] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Crescentic glomerulonephritis, a complication of severe immune glomerular injury, is the pathological correlate of rapidly progressive glomerulonephritis, mediated by both humoral and cellular effectors. In the current issue of the JCI, Chen et al. have implicated Bowman's capsule in functionally isolating potentially immune effectors, specifically antigen-specific CD8+ T lymphocytes, from podocytes. They suggest that, in crescentic glomerulonephritis, immune-mediated glomerular endothelial injury results in inside-out injury to the glomerulus, with subsequent leukocyte migration through a weakened or ruptured Bowman's capsule, resulting in outside-in injury. Effector T cells then recognize nephritogenic antigens presented by podocytes or other cells within the urinary space, enhancing injury and crescent formation.
Collapse
Affiliation(s)
- A Richard Kitching
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, Victoria, Australia.,Department of Nephrology and.,Department of Pediatric Nephrology, Monash Health, Clayton, Victoria, Australia
| | - Maliha A Alikhan
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, Victoria, Australia
| |
Collapse
|
13
|
Caster DJ, Korte EA, Tan M, Barati MT, Tandon S, Creed TM, Salant DJ, Hata JL, Epstein PN, Huang H, Powell DW, McLeish KR. Neutrophil exocytosis induces podocyte cytoskeletal reorganization and proteinuria in experimental glomerulonephritis. Am J Physiol Renal Physiol 2018; 315:F595-F606. [PMID: 29790391 DOI: 10.1152/ajprenal.00039.2018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Acute glomerulonephritis is characterized by rapid glomerular neutrophil recruitment, proteinuria, and glomerular hypercellularity. The current study tested the hypothesis that the release of neutrophil granule contents plays a role in both the loss of filtration barrier leading to proteinuria and the increase in glomerular cells. Inhibition of neutrophil exocytosis with a peptide inhibitor prevented proteinuria and attenuated podocyte and endothelial cell injury but had no effect on glomerular hypercellularity in an experimental acute glomerulonephritis model in mice. Cultivation of podocytes with neutrophil granule contents disrupted cytoskeletal organization, an in vitro model for podocyte effacement and loss of filtration barrier. Activated, cultured podocytes released cytokines that stimulated neutrophil chemotaxis, primed respiratory burst activity, and stimulated neutrophil exocytosis. We conclude that crosstalk between podocytes and neutrophils contributes to disruption of the glomerular filtration barrier in acute glomerulonephritis. Neutrophil granule products induce podocyte injury but do not participate in the proliferative response of intrinsic glomerular cells.
Collapse
Affiliation(s)
- Dawn J Caster
- Department of Medicine, University of Louisville , Louisville, Kentucky.,Robley Rex Veterans Affairs Medical Center , Louisville, Kentucky
| | - Erik A Korte
- Department of Biochemistry and Molecular Genetics, University of Louisville , Louisville, Kentucky
| | - Min Tan
- Department of Medicine, University of Louisville , Louisville, Kentucky
| | - Michelle T Barati
- Department of Medicine, University of Louisville , Louisville, Kentucky
| | - Shweta Tandon
- Department of Medicine, University of Louisville , Louisville, Kentucky
| | - T Michael Creed
- Department of Medicine, University of Louisville , Louisville, Kentucky
| | - David J Salant
- Department of Medicine, Boston University School of Medicine , Boston, Massachusetts
| | - Jessica L Hata
- Pathology Department, Norton Children's Hospital , Louisville, Kentucky
| | - Paul N Epstein
- Pediatric Research Institute in the Department of Pediatrics, University of Louisville , Louisville, Kentucky
| | - Hui Huang
- Pediatric Research Institute in the Department of Pediatrics, University of Louisville , Louisville, Kentucky.,Department of Endocrinology, Metabolism, and Genetics, Jiangxi Provincial Children's Hospital , Nanchang , China
| | - David W Powell
- Department of Medicine, University of Louisville , Louisville, Kentucky
| | - Kenneth R McLeish
- Department of Medicine, University of Louisville , Louisville, Kentucky.,Robley Rex Veterans Affairs Medical Center , Louisville, Kentucky
| |
Collapse
|
14
|
Hutton HL, Holdsworth SR, Kitching AR. ANCA-Associated Vasculitis: Pathogenesis, Models, and Preclinical Testing. Semin Nephrol 2018; 37:418-435. [PMID: 28863790 DOI: 10.1016/j.semnephrol.2017.05.016] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Our understanding of antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis has developed greatly since the discovery of ANCA, directed against neutrophil components, in 1982. Observations in human disease, and increasingly sophisticated studies in vitro and in rodent models in vivo, have allowed a nuanced understanding of many aspects of the immunopathogenesis of disease, including the significance of ANCA as a diagnostic and monitoring tool as well as a mediator of microvascular injury. The mechanisms of leukocyte recruitment and tissue injury, and the role of T cells increasingly are understood. Unexpected findings, such as the role of complement, also have been uncovered through experimental studies and human observations. This review focusses on the pathogenesis of ANCA-associated vasculitis, highlighting the challenges in finding new, less-toxic treatments and potential therapeutic targets in this disease. The current suite of rodent models is reviewed, and future directions in the study of this complex and fascinating disease are suggested.
Collapse
Affiliation(s)
- Holly L Hutton
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, Victoria, Australia; Department of Nephrology, Monash Health, Clayton, Victoria, Australia
| | - Stephen R Holdsworth
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, Victoria, Australia; Department of Nephrology, Monash Health, Clayton, Victoria, Australia
| | - A Richard Kitching
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, Victoria, Australia; Department of Nephrology, Monash Health, Clayton, Victoria, Australia; Department of Pediatric Nephrology, Monash Children's Hospital, Clayton, Victoria, Australia.
| |
Collapse
|
15
|
Alikhan MA, Huynh M, Kitching AR, Ooi JD. Regulatory T cells in renal disease. Clin Transl Immunology 2018; 7:e1004. [PMID: 29484182 PMCID: PMC5822411 DOI: 10.1002/cti2.1004] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 12/10/2017] [Accepted: 12/13/2017] [Indexed: 12/13/2022] Open
Abstract
The kidney is vulnerable to injury, both acute and chronic from a variety of immune and metabolic insults, all of which at least to some degree involve inflammation. Regulatory T cells modulate systemic autoimmune and allogenic responses in glomerulonephritis and transplantation. Intrarenal regulatory T cells (Tregs), including those recruited to the kidney, have suppressive effects on both adaptive and innate immune cells, and probably also intrinsic kidney cells. Evidence from autoimmune glomerulonephritis implicates antigen-specific Tregs in HLA-mediated dominant protection, while in several human renal diseases Tregs are abnormal in number or phenotype. Experimentally, Tregs can protect the kidney from injury in a variety of renal diseases. Mechanisms of Treg recruitment to the kidney include via the chemokine receptors CCR6 and CXCR3 and potentially, at least in innate injury TLR9. The effects of Tregs may be context dependent, with evidence for roles for immunoregulatory roles both for endogenous Tbet-expressing Tregs and STAT-3-expressing Tregs in experimental glomerulonephritis. Most experimental work and some of the ongoing human trials in renal transplantation have focussed on unfractionated thymically derived Tregs (tTregs). However, induced Tregs (iTregs), type 1 regulatory T (Tr1) cells and in particular antigen-specific Tregs also have therapeutic potential not only in renal transplantation, but also in other kidney diseases.
Collapse
Affiliation(s)
- Maliha A Alikhan
- Centre for Inflammatory Diseases Department of Medicine Monash University Monash Medical Centre Clayton Victoria Australia
| | - Megan Huynh
- Centre for Inflammatory Diseases Department of Medicine Monash University Monash Medical Centre Clayton Victoria Australia
| | - A Richard Kitching
- Centre for Inflammatory Diseases Department of Medicine Monash University Monash Medical Centre Clayton Victoria Australia.,Department of Nephrology Monash Health Clayton VIC Australia.,Department of Paediatric Nephrology Monash Health Clayton VIC Australia
| | - Joshua D Ooi
- Centre for Inflammatory Diseases Department of Medicine Monash University Monash Medical Centre Clayton Victoria Australia
| |
Collapse
|
16
|
Abstract
Inflammasomes influence a diverse range of kidney disease, including acute and chronic kidney diseases, and those mediated by innate and adaptive immunity. Both IL-18 and in particular IL-1β are validated therapeutic targets in several kidney diseases. In addition to leukocyte-derived inflammasomes, renal tissue cells express functional inflammasome components. Furthermore, a range of endogenous substances that directly activate inflammasomes also mediate kidney injury. Many of the functional studies have focussed on the NLRP3 inflammasome, and there is also evidence for the involvement of other inflammasomes in some conditions. While, at least in some disease, the mechanistic details of the involvement of the inflammasome remain to be elucidated, therapies focussed on inflammasomes and their products have potential in treating kidney disease in the future.
Collapse
Affiliation(s)
- Holly L Hutton
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, VIC, Australia
| | - Maliha A Alikhan
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, VIC, Australia
| | - A Richard Kitching
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, VIC, Australia.
- Department of Nephrology, Monash Health, Clayton, VIC, Australia.
- Department of Paediatric Nephrology, Monash Health, Clayton, VIC, Australia.
| |
Collapse
|
17
|
Affiliation(s)
- A Richard Kitching
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, Victoria, Australia; and .,Departments of Nephrology and.,Pediatric Nephrology, Monash Health, Clayton, Victoria, Australia
| | - Joshua D Ooi
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, Victoria, Australia; and
| |
Collapse
|
18
|
Korte EA, Caster DJ, Barati MT, Tan M, Zheng S, Berthier CC, Brosius FC, Vieyra MB, Sheehan RM, Kosiewicz M, Wysoczynski M, Gaffney PM, Salant DJ, McLeish KR, Powell DW. ABIN1 Determines Severity of Glomerulonephritis via Activation of Intrinsic Glomerular Inflammation. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:2799-2810. [PMID: 28935578 PMCID: PMC5718094 DOI: 10.1016/j.ajpath.2017.08.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 07/15/2017] [Accepted: 08/17/2017] [Indexed: 10/24/2022]
Abstract
Transcription factor NF-κB regulates expression of numerous genes that control inflammation and is activated in glomerular cells in glomerulonephritis (GN). We previously identified genetic variants for a NF-κB regulatory, ubiquitin-binding protein ABIN1 as risk factors for GN in systemic autoimmunity. The goal was to define glomerular inflammatory events controlled by ABIN1 function in GN. Nephrotoxic serum nephritis was induced in wild-type (WT) and ubiquitin-binding deficient ABIN1[D485N] mice, and renal pathophysiology and glomerular inflammatory phenotypes were assessed. Proteinuria was also measured in ABIN1[D485N] mice transplanted with WT mouse bone marrow. Inflammatory activation of ABIN1[D472N] (D485N homolog) cultured human-derived podocytes, and interaction with primary human neutrophils were also assessed. Disruption of ABIN1 function exacerbated proteinuria, podocyte injury, glomerular NF-κB activity, glomerular expression of inflammatory mediators, and glomerular recruitment and retention of neutrophils in antibody-mediated nephritis. Transplantation of WT bone marrow did not prevent the increased proteinuria in ABIN1[D845N] mice. Tumor necrosis factor-stimulated enhanced expression and secretion of NF-κB-targeted proinflammatory mediators in ABIN1[D472N] cultured podocytes compared with WT cells. Supernatants from ABIN1[D472N] podocytes accelerated chemotaxis of human neutrophils, and ABIN1[D472N] podocytes displayed a greater susceptibility to injurious morphologic findings induced by neutrophil granule contents. These studies define a novel role for ABIN1 dysfunction and NF-κB in mediating GN through proinflammatory activation of podocytes.
Collapse
Affiliation(s)
- Erik A Korte
- Department of Medicine University of Louisville, Louisville, Kentucky
| | - Dawn J Caster
- Department of Medicine University of Louisville, Louisville, Kentucky; Robley Rex VA Medical Center, Louisville, Kentucky
| | - Michelle T Barati
- Department of Medicine University of Louisville, Louisville, Kentucky
| | - Min Tan
- Department of Medicine University of Louisville, Louisville, Kentucky
| | - Shirong Zheng
- Department of Medicine University of Louisville, Louisville, Kentucky
| | - Celine C Berthier
- Department of Internal Medicine-Nephrology, University of Michigan, Ann Arbor, Michigan
| | - Frank C Brosius
- Department of Internal Medicine-Nephrology, University of Michigan, Ann Arbor, Michigan
| | - Mark B Vieyra
- Department of Medicine University of Louisville, Louisville, Kentucky
| | - Ryan M Sheehan
- Department of Medicine University of Louisville, Louisville, Kentucky
| | - Michele Kosiewicz
- Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky
| | | | - Patrick M Gaffney
- Arthritis and Clinical Immunology Program and Clinical Pharmacology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
| | - David J Salant
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
| | - Kenneth R McLeish
- Department of Medicine University of Louisville, Louisville, Kentucky; Department of Internal Medicine-Nephrology, University of Michigan, Ann Arbor, Michigan
| | - David W Powell
- Department of Medicine University of Louisville, Louisville, Kentucky.
| |
Collapse
|
19
|
Gan PY, Fujita T, Ooi JD, Alikhan MA, Dick J, Shim R, Odobasic D, O'Sullivan KM, Kitching AR, Holdsworth SR. Pathogenic Role for γδ T Cells in Autoimmune Anti-Myeloperoxidase Glomerulonephritis. THE JOURNAL OF IMMUNOLOGY 2017; 199:3042-3050. [PMID: 28954887 DOI: 10.4049/jimmunol.1602025] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 08/24/2017] [Indexed: 01/13/2023]
Abstract
Myeloperoxidase (MPO) anti-neutrophil cytoplasmic Ab (ANCA)-associated vasculitis results from autoimmunity to MPO. IL-17A plays a critical role in generating this form of autoimmune injury but its cell of origin is uncertain. We addressed the hypothesis that IL-17A-producing γδ T cells are a nonredundant requisite in the development of MPO autoimmunity and glomerulonephritis (GN). We studied MPO-ANCA GN in wild type, αβ, or γδ T cell-deficient (C57BL/6, βTCR-/- , and δTCR-/- respectively) mice. Both T cell populations played important roles in the generation of autoimmunity to MPO and GN. Humoral autoimmunity was dependent on intact αβ T cells but was unaffected by γδ T cell deletion. Following MPO immunization, activated γδ T cells migrate to draining lymph nodes. Studies in δTCR-/- and transfer of γδ T cells to δTCR-/- mice show that γδ T cells facilitate the generation of anti-MPO autoimmunity and GN. δTCR-/- mice that received IL-17A-/- γδ T cells demonstrate that the development of anti-MPO autoimmunity and GN are dependent on γδ T cell IL-17A production. Finally, transfer of anti-MPO CD4+ T cell clones to naive δTCR-/- and wild type mice with planted glomerular MPO shows that γδ T cells are also necessary for recruitment of anti-MPO αβ CD4+ effector T cells. This study demonstrates that IL-17A produced by γδ T cells plays a critical role in the pathogenesis of MPO-ANCA GN by promoting the development of MPO-specific αβ T cells.
Collapse
Affiliation(s)
- Poh-Yi Gan
- Centre for Inflammatory Diseases, Department of Medicine, Monash Medical Centre, Monash University, Clayton, Victoria 3168, Australia; .,Department of Immunology, Monash Health, Monash Medical Centre, Clayton, Victoria 3168, Australia
| | - Takeshi Fujita
- Department of Cardiology, Respiratory Medicine and Nephrology, Hirosaki University Graduate School of Medicine, Hirosaki 036-8562, Japan; and
| | - Joshua Daniel Ooi
- Centre for Inflammatory Diseases, Department of Medicine, Monash Medical Centre, Monash University, Clayton, Victoria 3168, Australia
| | - Maliha Asghar Alikhan
- Centre for Inflammatory Diseases, Department of Medicine, Monash Medical Centre, Monash University, Clayton, Victoria 3168, Australia
| | - Jonathan Dick
- Centre for Inflammatory Diseases, Department of Medicine, Monash Medical Centre, Monash University, Clayton, Victoria 3168, Australia.,Department of Nephrology, Monash Health, Monash Medical Centre, Clayton, Victoria 3168, Australia
| | - Raymond Shim
- Centre for Inflammatory Diseases, Department of Medicine, Monash Medical Centre, Monash University, Clayton, Victoria 3168, Australia
| | - Dragana Odobasic
- Centre for Inflammatory Diseases, Department of Medicine, Monash Medical Centre, Monash University, Clayton, Victoria 3168, Australia
| | - Kim Maree O'Sullivan
- Centre for Inflammatory Diseases, Department of Medicine, Monash Medical Centre, Monash University, Clayton, Victoria 3168, Australia
| | - Arthur Richard Kitching
- Centre for Inflammatory Diseases, Department of Medicine, Monash Medical Centre, Monash University, Clayton, Victoria 3168, Australia.,Department of Nephrology, Monash Health, Monash Medical Centre, Clayton, Victoria 3168, Australia
| | - Stephen Roger Holdsworth
- Centre for Inflammatory Diseases, Department of Medicine, Monash Medical Centre, Monash University, Clayton, Victoria 3168, Australia.,Department of Immunology, Monash Health, Monash Medical Centre, Clayton, Victoria 3168, Australia.,Department of Nephrology, Monash Health, Monash Medical Centre, Clayton, Victoria 3168, Australia
| |
Collapse
|
20
|
Ghali JR, Wang YM, Holdsworth SR, Kitching AR. Regulatory T cells in immune-mediated renal disease. Nephrology (Carlton) 2016. [PMID: 26206106 DOI: 10.1111/nep.12574] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Regulatory T cells (Tregs) are CD4+ T cells that can suppress immune responses by effector T cells, B cells and innate immune cells. This review discusses the role that Tregs play in murine models of immune-mediated renal diseases and acute kidney injury and in human autoimmune kidney disease (such as systemic lupus erythematosus, anti-glomerular basement membrane disease, anti-neutrophil cytoplasmic antibody-associated vasculitis). Current research suggests that Tregs may be reduced in number and/or have impaired regulatory function in these diseases. Tregs possess several mechanisms by which they can limit renal and systemic inflammatory immune responses. Potential therapeutic applications involving Tregs include in vivo induction of Tregs or inducing Tregs from naïve CD4+ T cells or expanding natural Tregs ex vivo, to use as a cellular therapy. At present, the optimal method of generating a phenotypically stable pool of Tregs with long-lasting suppressive effects is not established, but human studies in renal transplantation are underway exploring the therapeutic potential of Tregs as a cellular therapy, and if successful may have a role as a novel therapy in immune-mediated renal diseases.
Collapse
Affiliation(s)
- Joanna R Ghali
- Centre for Inflammatory Diseases, Department of Medicine, Monash University, Melbourne, Victoria.,Department of Nephrology, Monash Medical Centre, Melbourne, Victoria
| | - Yuan Min Wang
- Centre for Kidney Research, Children's Hospital at Westmead, The University of Sydney, Westmead, New South Wales, Australia
| | - Stephen R Holdsworth
- Centre for Inflammatory Diseases, Department of Medicine, Monash University, Melbourne, Victoria.,Department of Nephrology, Monash Medical Centre, Melbourne, Victoria
| | - A Richard Kitching
- Centre for Inflammatory Diseases, Department of Medicine, Monash University, Melbourne, Victoria.,Department of Nephrology, Monash Medical Centre, Melbourne, Victoria.,Department of Paediatric Nephrology, Monash Medical Centre, Melbourne, Victoria
| |
Collapse
|
21
|
Ghali JR, Alikhan MA, Holdsworth SR, Kitching AR. Induced regulatory T cells are phenotypically unstable and do not protect mice from rapidly progressive glomerulonephritis. Immunology 2016; 150:100-114. [PMID: 27606831 DOI: 10.1111/imm.12671] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 08/10/2016] [Accepted: 09/02/2016] [Indexed: 12/15/2022] Open
Abstract
Regulatory T (Treg) cells are a suppressive CD4+ T-cell subset. We generated induced Treg (iTreg) cells and explored their therapeutic potential in a murine model of rapidly progressive glomerulonephritis. Polyclonal naive CD4+ T cells were cultured in vitro with interleukin-2 (IL-2), transforming growth factor-β1, all-trans-retinoic acid and monoclonal antibodies against interferon-γ and IL-4, generating Foxp3+ iTreg cells. To enhance their suppressive phenotype, iTreg cultures were modified with the addition of a monoclonal antibody against IL-12p40 or by using RORγt-/- CD4+ T cells. Induced Treg cells were transferred into models of delayed-type hypersensitivity and experimental glomerulonephritis. The iTreg cells exhibited comparable surface receptor expression and in vitro suppressive ability to natural Treg cells, but did not regulate antigen-specific delayed-type hypersensitivity or systemic inflammatory immune responses, losing Foxp3 expression in vivo. In glomerulonephritis, transferred iTreg cells did not prevent renal injury or modulate systemic T helper type 1 immune responses. Induced Treg cells cultured with anti-IL-12p40 had an enhanced suppressive phenotype in vitro and regulated dermal delayed-type hypersensitivity in vivo, but were not protective against renal injury, losing Foxp3 expression, especially in the transferred cells recruited to the kidney. Use of RORγt-/- CD4+ T cells or iTreg cells generated from sensitized CD4+ Foxp3- cells did not regulate renal or systemic inflammatory responses in vivo. In conclusion, iTreg cells suppress T-cell proliferation in vitro, but do not regulate experimental glomerulonephritis, being unstable in this inflammatory milieu in vivo.
Collapse
Affiliation(s)
- Joanna R Ghali
- Centre for Inflammatory Diseases, Department of Medicine, Monash University, Clayton, Victoria, Australia.,Department of Nephrology, Monash Health, Clayton, Victoria, Australia
| | - Maliha A Alikhan
- Centre for Inflammatory Diseases, Department of Medicine, Monash University, Clayton, Victoria, Australia
| | - Stephen R Holdsworth
- Centre for Inflammatory Diseases, Department of Medicine, Monash University, Clayton, Victoria, Australia.,Department of Nephrology, Monash Health, Clayton, Victoria, Australia
| | - A Richard Kitching
- Centre for Inflammatory Diseases, Department of Medicine, Monash University, Clayton, Victoria, Australia.,Department of Nephrology, Monash Health, Clayton, Victoria, Australia.,Department of Paediatric Nephrology, Monash Health, Clayton, Victoria, Australia
| |
Collapse
|
22
|
Abstract
Biological therapeutics (biologics) that target autoimmune responses and inflammatory injury pathways have a marked beneficial impact on the management of many chronic diseases, including rheumatoid arthritis, psoriasis, inflammatory bowel disease, and ankylosing spondylitis. Accumulating data suggest that a growing number of renal diseases result from autoimmune injury - including lupus nephritis, IgA nephropathy, anti-neutrophil cytoplasmic antibody-associated glomerulonephritis, autoimmune (formerly idiopathic) membranous nephropathy, anti-glomerular basement membrane glomerulonephritis, and C3 nephropathy - and one can speculate that biologics might also be applicable to these diseases. As many autoimmune renal diseases are relatively uncommon, with long natural histories and diverse outcomes, clinical trials that aim to validate potentially useful biologics are difficult to design and/or perform. Some excellent consortia are undertaking cohort studies and clinical trials, but more multicentre international collaborations are needed to advance the introduction of new biologics to patients with autoimmune renal disorders. This Review discusses the key molecules that direct injurious inflammation and the biologics that are available to modulate them. The opportunities and challenges for the introduction of relevant biologics into treatment protocols for autoimmune renal diseases are also discussed.
Collapse
|