23 results on '"Giulia Antonelli"'
Search Results
2. Preparation of Human Kidney Progenitor Cultures and Their Differentiation into Podocytes
- Author
-
Maria Elena Melica, Maria Lucia Angelotti, Giulia Antonelli, Anna Peired, Carolina Conte, Letizia De Chiara, Benedetta Mazzinghi, Elena Lazzeri, Laura Lasagni, and Paola Romagnani
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Kidney diseases are a global health concern. Modeling of kidney disease for translational research is often challenging because of species specificities or the postmitotic status of kidney epithelial cells that make primary cultures, for example podocytes. Here, we report a protocol for preparing primary cultures of podocytes based on the isolation and in vitro propagation of immature kidney progenitor cells subsequently differentiated into mature podocytes. This protocol can be useful for studying physiology and pathophysiology of human kidney progenitors and to obtain differentiated podocytes for modeling podocytopathies and other kidney disorders involving podocytes.Graphical overview
- Published
- 2023
- Full Text
- View/download PDF
3. No NLRP3 inflammasome activity in kidney epithelial cells, not even when the NLRP3-A350V Muckle-Wells variant is expressed in podocytes of diabetic mice
- Author
-
Sophie Carina Kunte, Julian A. Marschner, Martin Klaus, Tâmisa Honda, Chenyu Li, Manga Motrapu, Christoph Walz, Maria Lucia Angelotti, Giulia Antonelli, Maria Elena Melica, Letizia De Chiara, Roberto Semeraro, Peter J. Nelson, and Hans-Joachim Anders
- Subjects
inflammasome ,inflammation ,diabetes ,chronic kidney disease ,IL-1 ,proteinuria ,Immunologic diseases. Allergy ,RC581-607 - Abstract
BackgroundThe NLRP3 inflammasome integrates several danger signals into the activation of innate immunity and inflammation by secreting IL-1β and IL-18. Most published data relate to the NLRP3 inflammasome in immune cells, but some reports claim similar roles in parenchymal, namely epithelial, cells. For example, podocytes, epithelial cells critical for the maintenance of kidney filtration, have been reported to express NLRP3 and to release IL-β in diabetic kidney disease, contributing to filtration barrier dysfunction and kidney injury. We questioned this and hence performed independent verification experiments.MethodsWe studied the expression of inflammasome components in human and mouse kidneys and human podocytes using single-cell transcriptome analysis. Human podocytes were exposed to NLRP3 inflammasome agonists in vitro and we induced diabetes in mice with a podocyte-specific expression of the Muckle-Wells variant of NLRP3, leading to overactivation of the Nlrp3 inflammasome (Nphs2Cre;Nlrp3A350V) versus wildtype controls. Phenotype analysis included deep learning-based glomerular and podocyte morphometry, tissue clearing, and STED microscopy of the glomerular filtration barrier. The Nlrp3 inflammasome was blocked by feeding ß-hydroxy-butyrate.ResultsSingle-cell transcriptome analysis did not support relevant NLRP3 expression in parenchymal cells of the kidney. The same applied to primary human podocytes in which NLRP3 agonists did not induce IL-1β or IL-18 secretion. Diabetes induced identical glomerulomegaly in wildtype and Nphs2Cre;Nlrp3A350V mice but hyperfiltration-induced podocyte loss was attenuated and podocytes were larger in Nphs2Cre;Nlrp3A350V mice, an effect reversible with feeding the NLRP3 inflammasome antagonist ß-hydroxy-butyrate. Ultrastructural analysis of the slit diaphragm was genotype-independent hence albuminuria was identical.ConclusionPodocytes express low amounts of the NLRP3 inflammasome, if at all, and do not produce IL-1β and IL-18, not even upon introduction of the A350V Muckle-Wells NLRP3 variant and upon induction of podocyte stress. NLRP3-mediated glomerular inflammation is limited to immune cells.
- Published
- 2023
- Full Text
- View/download PDF
4. DRD2, DRD3, and HTR2A Single-Nucleotide Polymorphisms Involvement in High Treatment Resistance to Atypical Antipsychotic Drugs
- Author
-
Antonio Del Casale, Maurizio Simmaco, Martina Nicole Modesti, Clarissa Zocchi, Jan Francesco Arena, Irene Bilotta, Alessandro Alcibiade, Giuseppe Sarli, Lorenzo Cutillo, Giulia Antonelli, Enrico La Spina, Ottavia De Luca, Robert Preissner, Marina Borro, Giovanna Gentile, Paolo Girardi, and Maurizio Pompili
- Subjects
antipsychotic agents ,dopamine receptors ,5-hydroxytryptamine receptors ,drug resistance ,pharmacogenomic variants ,precision medicine ,Biology (General) ,QH301-705.5 - Abstract
Background: The objective of this study was to investigate the DRD2 rs1800497, rs1799732, rs1801028, DRD3 rs6280, and HTR2A rs6314, rs7997012, and rs6311 single-nucleotide polymorphism (SNP) correlations with resistance to second-generation antipsychotics (SGAs) in a real-world sample of patients with treatment-resistant mental disorders. Methods: We divided 129 participants into a high treatment resistance (HTR) group (current treatment with two SGAs, or clozapine, or classic neuroleptics for a failure of previous SGAs trials) and a low treatment resistance (LTR) group (current treatment with one atypical antipsychotic). We used Next-Generation Sequencing on DNA isolated from peripheral blood samples to analyze the polymorphisms. We performed logistic regression to search for predictors of HTR membership. Results: A diagnosis of schizophrenia significantly predicted the HTR membership compared to other diagnoses. Other predictors were the DRD3 rs6280 C|T (OR = 22.195) and T|T (OR = 18.47) vs. C|C, HTR2A rs7997012 A|G vs. A|A (OR = 6.859) and vs. G|G (OR = 2.879), and DRD2 rs1799732 I|I vs. D|I (OR = 12.079) genotypes. Conclusions: A diagnosis of schizophrenia and the DRD2 rs1799732, DRD3 rs6280, and HTR2A rs7997012 genotypes can predict high treatment resistance to SGAs.
- Published
- 2023
- Full Text
- View/download PDF
5. Endocycle-related tubular cell hypertrophy and progenitor proliferation recover renal function after acute kidney injury
- Author
-
Elena Lazzeri, Maria Lucia Angelotti, Anna Peired, Carolina Conte, Julian A. Marschner, Laura Maggi, Benedetta Mazzinghi, Duccio Lombardi, Maria Elena Melica, Sara Nardi, Elisa Ronconi, Alessandro Sisti, Giulia Antonelli, Francesca Becherucci, Letizia De Chiara, Ricardo Romero Guevara, Alexa Burger, Beat Schaefer, Francesco Annunziato, Hans-Joachim Anders, Laura Lasagni, and Paola Romagnani
- Subjects
Science - Abstract
The recovery of function upon acute kidney injury is thought to involve tubular cell dedifferentiation and proliferation. Here the authors show that Pax2+ progenitors regenerate tubules via cell division while other tubular cells support function recovery by undergoing hypertrophy through endoreplication.
- Published
- 2018
- Full Text
- View/download PDF
6. Differentiation of crescent-forming kidney progenitor cells into podocytes attenuates severe glomerulonephritis in mice
- Author
-
Maria Elena Melica, Giulia Antonelli, Roberto Semeraro, Maria Lucia Angelotti, Gianmarco Lugli, Samuela Landini, Fiammetta Ravaglia, Gilda La Regina, Carolina Conte, Letizia De Chiara, Anna Julie Peired, Benedetta Mazzinghi, Marta Donati, Alice Molli, Stefanie Steiger, Alberto Magi, Niccolò Bartalucci, Valentina Raglianti, Francesco Guzzi, Laura Maggi, Francesco Annunziato, Alexa Burger, Elena Lazzeri, Hans-Joachim Anders, Laura Lasagni, and Paola Romagnani
- Subjects
Disease Models, Animal ,Mice ,Glomerulonephritis ,Podocytes ,Stem Cells ,Panobinostat ,Animals ,Humans ,General Medicine ,Kidney ,Article - 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.
- Published
- 2022
7. FC071: An HDAC Inhibitor Attenuates Crescentic Glomerulonephritis and Avoid Chronic Kidney Disease Enhancing Podocyte Progenitor Differentiation
- Author
-
Maria Elena Melica, Giulia Antonelli, Roberto Semeraro, Maria Lucia Angelotti, Letizia De Chiara, Carolina Conte, Gianmarco Lugli, Anna Julie Peired, Gilda La Regina, Benedetta Mazzinghi, Elena Lazzeri, Laura Lasagni, and Paola Romagnani
- Subjects
Transplantation ,Nephrology - Abstract
BACKGROUND AND AIMS Crescentic glomerulonephritis (crescentic GN) encompasses a group of diverse disorders characterized by the presence of massive hyperplasia of parietal epithelial cells (PEC) as the main histopathological lesion at kidney biopsy. It is associated with a rapid decline in kidney function. Typically, crescent formation is the consequence of diverse upstream pathomechanisms involving the specific activation of PEC. PEC normally reside peacefully along Bowman capsule and represent in part renal progenitor cells (RPC). Previous studies observed RPC markers in crescents from patients with different types of glomerulonephritis. Similarities between stem cell niches of bone marrow and kidney, prompted us to hypothesized that crescents result from monoclonal expansion of a single RPC clone conceptually similar to monoclonal diseases originating from hematopoietic stem cells. According to this analogy, we further hypothesized that drugs known to cure monoclonal disease of the hematopoietic stem cells by enforcing their terminal differentiation could also attenuate crescentic glomerulonephritis. METHOD To address this hypothesis, we established a crescentic GN disease model in a conditional transgenic mouse based on the mT/mG and the Confetti reporter that allows lineage tracing and clonal analysis of RPCs. Animals were treated with known pharmacological inhibitors of clonal stem cell proliferation in myeloproliferative disorders. Crescentic lesions were characterized by super-resolution STED microscopy. Finally, we employed single cell RNA sequencing of human renal progenitor cultures to identify the immature progenitor subset-generating crescent in human to identify putative new biomarkers of crescentic GN to validate in biopsy of patients. RESULTS We observed that the crescentic lesions originated from the clonal expansion of single RPC, thus suggesting a clonal stem cell disorder. Therefore, we administrated a series of drugs known to ameliorates myeloproliferative neoplasms to our crescentic GN mouse model as potential therapeutic agents. Treatment with one of the compounds induced a reduction in both proteinuria and crescent formation. STED super-resolution imaging of glomeruli showed that this compound turned the uncontrolled hyperplasia of a specific immature PEC subset into a controlled differentiation into podocytes thereby restoring the injured glomerular filtration barrier. Moreover, delayed drug administration still induced proteinuria remission and avoided long-term development of chronic kidney disease (CKD), an effect associated to a continued generation of new podocytes and crescent regression over time. Single cell RNA sequencing of human RPC identified a new marker of the crescent-generating progenitor cells. Expression of this marker in biopsies of patients with crescentic GN associated with progression toward end stage kidney disease. Treatment of human PEC with the drug that in in vivo experiments showed a therapeutic effect on crescentic GN reduced proliferation of the immature progenitor subset promoting their differentiation into podocytes. CONCLUSION These results demonstrate that glomerular hyperplastic lesions derive from clonal amplification of a RPC subset and that shifting proliferation to podocyte differentiation reverses improves clinical outcome and avoid CKD.
- Published
- 2022
- Full Text
- View/download PDF
8. Drug Testing for Residual Progression of Diabetic Kidney Disease in Mice Beyond Therapy with Metformin, Ramipril, and Empagliflozin
- Author
-
Yutian Lei, Hans-Joachim Anders, Giulia Antonelli, Maria Lucia Angelotti, Laura Martinez Valenzuela, Manga Motrapu, Julian A. Marschner, Lidia Anguiano, Irene Mesas, Monika Katarzyna Świderska, Paola Romagnani, Kyung Lee, and Jia Fu
- Subjects
Ramipril ,medicine.medical_specialty ,Indoles ,Combination therapy ,030232 urology & nephrology ,Urology ,Type 2 diabetes ,Kidney ,Diabetic nephropathy ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Glucosides ,Oximes ,medicine ,Empagliflozin ,Animals ,Diabetic Nephropathies ,Benzhydryl Compounds ,030304 developmental biology ,0303 health sciences ,Glycogen Synthase Kinase 3 beta ,Podocytes ,business.industry ,Glomerulosclerosis ,General Medicine ,medicine.disease ,Metformin ,3. Good health ,Basic Research ,Diabetes Mellitus, Type 2 ,Nephrology ,Disease Progression ,Albuminuria ,Drug Therapy, Combination ,medicine.symptom ,business ,Glomerular Filtration Rate ,medicine.drug - Abstract
Background Progression of CKD in type 2 diabetes, despite dual inhibition of sodium-glucose transporter-2 and the renin-angiotensin system, remains a concern. Bromoindirubin-3'-oxime (BIO), previously reported to promote podocyte survival and regeneration, is a candidate additional drug to elicit renoprotective effects beyond therapy with metformin, ramipril, and empagliflozin (MRE). Evaluating a drug with standard therapeutics more closely mimics the clinical setting than evaluating the drug alone. Methods Uninephrectomized BKS-Lepr-/- (db/db) mice treated with or without MRE served as a model of progressive CKD in type 2 diabetes. Mice on or off MRE were randomized to only 4 weeks of add-on BIO or vehicle. The primary end point was slope of GFR (ΔGFR). Results Four weeks of MRE treatment alone did not affect ΔGFR, but significantly attenuated hyperglycemia, albuminuria, and glomerulosclerosis and increased podocyte filtration slit density, as assessed by STED super-resolution microscopy upon tissue clearing. BIO alone improved albuminuria, podocyte density in superficial and juxtamedullary nephrons, and podocyte filtration slit density. MRE+BIO combination therapy had additive protective effects on ΔGFR, glomerulosclerosis, podocyte density in juxtamedullary nephrons, and filtration slit density. Conclusions Add-on treatment with BIO for only 4 weeks attenuates progression of CKD beyond MRE therapy in mice with type 2 diabetes. Additional drug combinations may help to further delay ESKD in type 2 diabetes.
- Published
- 2020
- Full Text
- View/download PDF
9. Tubular Cell Cycle Response upon AKI: Revising Old and New Paradigms to Identify Novel Targets for CKD Prevention
- Author
-
Letizia De Chiara, Carolina Conte, Elena Lazzeri, and Giulia Antonelli
- Subjects
senescence ,030232 urology & nephrology ,Review ,urologic and male genital diseases ,0302 clinical medicine ,Mitotic cell cycle ,Fibrosis ,Molecular Targeted Therapy ,Biology (General) ,Spectroscopy ,polyploidy ,0303 health sciences ,Kidney ,Cell Cycle ,Acute kidney injury ,General Medicine ,Acute Kidney Injury ,female genital diseases and pregnancy complications ,3. Good health ,Computer Science Applications ,Chemistry ,medicine.anatomical_structure ,Kidney Tubules ,cell cycle arrest ,Senescence ,medicine.medical_specialty ,QH301-705.5 ,Renal function ,Mitosis ,mitotic cell cycle ,Catalysis ,Inorganic Chemistry ,03 medical and health sciences ,alternative cell cycle ,medicine ,Animals ,Humans ,Cell Lineage ,Physical and Theoretical Chemistry ,Renal Insufficiency, Chronic ,Intensive care medicine ,Molecular Biology ,QD1-999 ,Disease burden ,030304 developmental biology ,business.industry ,urogenital system ,Organic Chemistry ,fibrosis ,Cell Cycle Checkpoints ,medicine.disease ,Histone Deacetylase Inhibitors ,acute kidney injury ,chronic kidney disease ,business ,Kidney disease - Abstract
Acute kidney injury (AKI) is characterized by a rapid deterioration of kidney function, representing a global healthcare concern. In addition, AKI survivors frequently develop chronic kidney disease (CKD), contributing to a substantial proportion of disease burden globally. Yet, over the past 30 years, the burden of CKD has not declined to the same extent as many other important non-communicable diseases, implying a substantial deficit in the understanding of the disease progression. The assumption that the kidney response to AKI is based on a high proliferative potential of proximal tubular cells (PTC) caused a critical confounding factor, which has led to a limited development of strategies to prevent AKI and halt progression toward CKD. In this review, we discuss the latest findings on multiple mechanisms of response related to cell cycle behavior of PTC upon AKI, with a specific focus on their biological relevance. Collectively, we aim to (1) provide a new perspective on interpreting cell cycle progression of PTC in response to damage and (2) discuss how this knowledge can be used to choose the right therapeutic window of treatment for preserving kidney function while avoiding CKD progression.
- Published
- 2021
10. Tubular cell polyploidy protects from lethal acute kidney injury but promotes consequent chronic kidney disease
- Author
-
Letizia De Chiara, Carolina Conte, Roberto Semeraro, Paula Diaz-Bulnes, Maria Lucia Angelotti, Benedetta Mazzinghi, Alice Molli, Giulia Antonelli, Samuela Landini, Maria Elena Melica, Anna Julie Peired, Laura Maggi, Marta Donati, Gilda La Regina, Marco Allinovi, Fiammetta Ravaglia, Daniele Guasti, Daniele Bani, Luigi Cirillo, Francesca Becherucci, Francesco Guzzi, Alberto Magi, Francesco Annunziato, Laura Lasagni, Hans-Joachim Anders, Elena Lazzeri, and Paola Romagnani
- Subjects
Polyploidy ,Multidisciplinary ,Senotherapeutics ,Disease Progression ,General Physics and Astronomy ,Humans ,RNA ,General Chemistry ,DNA ,Acute Kidney Injury ,Renal Insufficiency, Chronic ,Kidney ,General Biochemistry, Genetics and Molecular Biology - Abstract
Acute kidney injury (AKI) is frequent, often fatal and, for lack of specific therapies, can leave survivors with chronic kidney disease (CKD). We characterize the distribution of tubular cells (TC) undergoing polyploidy along AKI by DNA content analysis and single cell RNA-sequencing. Furthermore, we study the functional roles of polyploidization using transgenic models and drug interventions. We identify YAP1-driven TC polyploidization outside the site of injury as a rapid way to sustain residual kidney function early during AKI. This survival mechanism comes at the cost of senescence of polyploid TC promoting interstitial fibrosis and CKD in AKI survivors. However, targeting TC polyploidization after the early AKI phase can prevent AKI-CKD transition without influencing AKI lethality. Senolytic treatment prevents CKD by blocking repeated TC polyploidization cycles. These results revise the current pathophysiological concept of how the kidney responds to acute injury and identify a novel druggable target to improve prognosis in AKI survivors.
- Published
- 2021
11. FC 038CRESCENTS DERIVE FROM SINGLE PODOCYTE PROGENITORS AND A DRUG ENHANCING THEIR DIFFERENTIATION ATTENUATES RAPIDLY PROGRESSIVE GLOMERULONEPHRITIS
- Author
-
Paola Romagnani, Giulia Antonelli, Maria Elena Melica, Maria Lucia Angelotti, Laura Lasagni, Roberto Semeraro, Gianmarco Lugli, and Elena Lazzeri
- Subjects
Drug ,Transplantation ,Pathology ,medicine.medical_specialty ,Proteinuria ,medicine.diagnostic_test ,business.industry ,media_common.quotation_subject ,Myeloproliferative disease ,medicine.disease ,Podocyte ,medicine.anatomical_structure ,Nephrology ,medicine ,Rapidly progressive glomerulonephritis ,Renal biopsy ,Progenitor cell ,medicine.symptom ,Stem cell ,business ,media_common - Abstract
Background and Aims Rapidly progressive glomerulonephritis (RPGN) encompasses a group of diverse disorders characterized by the presence of massive hyperplasia of parietal epithelial cells (PEC) as the main histopathological lesion at kidney biopsy. It is associated with a rapid decline in kidney function referred to altogether as rapidly progressive glomerulonephritis. Typically, crescent formation is the consequence of diverse upstream pathomechanisms involving the specific activation of PEC. PEC normally reside peacefully along Bowman capsule and represent in part renal progenitor cells (RPC). Previous studies observed RPC markers in crescents from patients with different types of glomerulonephritis. Similarities between stem cell niches of bone marrow and kidney, prompted us to hypothesized that crescents result from monoclonal expansion of a single RPC clone conceptually similar to monoclonal diseases originating from hematopoietic stem cells. According to this analogy, we further hypothesized that drugs known to cure monoclonal disease of the hematopoietic stem cells by enforcing their terminal differentiation could also attenuate crescentic glomerulonephritis. Method To address this hypothesis, we established a RPGN disease model in a conditional transgenic mouse based on the mT/mG and the Confetti reporter that allows lineage tracing and clonal analysis of RPCs. Animals were treated with known pharmacological inhibitors of clonal stem cell proliferation in myeloproliferative disorders. Crescentic lesions were characterized by super-resolution STED microscopy. Finally, we employed single cell RNA sequencing of human renal progenitor cultures to identify the immature progenitor subset-generating crescent in human to identify putative new biomarker(s) of RPNG to validate in biopsy of patients. Results We observed that the crescentic lesions originated from the clonal expansion of single RPC, thus suggesting a clonal stem cell disorder. Therefore, we administrated a series of drugs known to ameliorates myeloproliferative neoplasms to our RPGN mouse model as potential therapeutic agents. In particular, treatment with one of the compounds induced a reduction in both proteinuria and crescent formation. 3D confocal microscopy and STED super-resolution imaging of glomeruli showed that this compound turned the uncontrolled hyperplasia of a specific immature PEC subset into a controlled differentiation into new podocytes thereby restoring the injured glomerular filtration barrier. Single cell RNA sequencing of human renal progenitor cultures identified a new marker of the crescent-generating progenitor cells. Expression of this marker in biopsies of patients with rapidly progressive glomerulonephritis associated with progression toward end stage kidney disease. Treatment of human PEC with the drug that in in vivo experiments showed a therapeutic effect on RPGN reduced proliferation of the immature progenitor subset promoting their differentiation into podocytes. Conclusion These results demonstrate that glomerular hyperplastic lesions derive from clonal amplification of a RPC subset and that shifting proliferation to podocyte differentiation reverses crescent formation and improves clinical outcome.
- Published
- 2021
- Full Text
- View/download PDF
12. Sex and Gender Differences in Kidney Cancer: Clinical and Experimental Evidence
- Author
-
Francesca Becherucci, Linda Calistri, Sergio Serni, Carolina Conte, Riccardo Campi, Anna Julie Peired, Elena Lazzeri, Paola Romagnani, Maria Lucia Angelotti, and Giulia Antonelli
- Subjects
renal cell carcinoma ,Cancer Research ,Female sex hormones ,Review ,outcomes ,Bioinformatics ,Diagnostic tools ,hormone signaling axis ,Renal cell carcinoma ,gender ,medicine ,sex ,risk factors ,RC254-282 ,business.industry ,Incidence (epidemiology) ,Mortality rate ,kidney cancer ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,Obesity ,Oncology ,incidence ,business ,Kidney cancer ,Hormone - Abstract
Simple Summary Kidney cancer is a frequent malignant tumor that accounts for approximately 5% of all cancer incidences. It affects both males and females, but males are twice as likely to develop kidney cancer than females. Evidence shows that this discrepancy takes root in individual differences, such as genetics or pathologies that affect the patient. It is then reflected in the clinical characteristics of the tumors, as males have larger and more aggressive tumors. Understanding the sex- and gender-based differences in kidney cancer is essential to be able to offer patients individualized medicine that would better cover their needs in terms of prevention, diagnosis and treatment. Abstract Sex and gender disparities have been reported for different types of non-reproductive cancers. Males are two times more likely to develop kidney cancer than females and have a higher death rate. These differences can be explained by looking at genetics and genomics, as well as other risk factors such as hypertension and obesity, lifestyle, and female sex hormones. Examination of the hormonal signaling pathways bring further insights into sex-related differences. Sex and gender-based disparities can be observed at the diagnostic, histological and treatment levels, leading to significant outcome difference. This review summarizes the current knowledge about sex and gender-related differences in the clinical presentation of patients with kidney cancer and the possible biological mechanisms that could explain these observations. Underlying sex-based differences may contribute to the development of sex-specific prognostic and diagnostic tools and the improvement of personalized therapies.
- Published
- 2021
- Full Text
- View/download PDF
13. Collapsing Glomerulopathy as a Complication of Type I Interferon–Mediated Glomerulopathy in a Patient With RNASEH2B-Related Aicardi-Goutières Syndrome
- Author
-
Paola Romagnani, Giovanni Maria Rossi, Augusto Vaglio, Maria Lucia Angelotti, Stefano Volpi, Lorenzo Lodi, Samuela Landini, Giulia Antonelli, Paride Fenaroli, Alice Grossi, and Marco Delsante
- Subjects
Pathology ,medicine.medical_specialty ,kidney biopsy ,030232 urology & nephrology ,case report ,Collapsing glomerulopathy (CG) ,genetic mutation ,interferon ,parietal epithelial cells ,RNASEH2B ,systemic lupus erythematosus (SLE) ,03 medical and health sciences ,0302 clinical medicine ,Membranous nephropathy ,Interferon ,Glomerulopathy ,Biopsy ,medicine ,030212 general & internal medicine ,Kidney ,medicine.diagnostic_test ,biology ,business.industry ,CD44 ,medicine.disease ,medicine.anatomical_structure ,Nephrology ,biology.protein ,Aicardi–Goutières syndrome ,Synaptopodin ,business ,medicine.drug - Abstract
Aicardi-Goutieres syndrome (AGS) is a well-characterized monogenic type I interferonopathy presenting with prominent neurologic manifestations. Among extraneurologic features, renal involvement has been described in only 1 patient with an IFIH1 mutation in whom membranous nephropathy developed. The pathogenic role of augmented interferon (IFN) signaling in tissues other than the central nervous system remains to be elucidated. We report a case of collapsing glomerulopathy in a 15-year-old girl affected by AGS with RNASEH2B mutation (an alanine-to-threonine change at amino acid 177), which led to kidney failure. The patient had no lupus-like features and lacked the APOL1 G1 and G2 risk alleles. Kidney biopsy showed findings consistent with collapsing glomerulopathy. MxA, a protein involved in antiviral immunity and induced by type I IFNs, was selectively expressed in CD133-positive parietal epithelial cells (PECs) but not in podocytes that stained for synaptopodin or in other glomerular cells. MxA also colocalized within pseudocrescents with CD44, a marker of PEC activation involved in cellular proliferation, differentiation, and migration and in glomerular scarring. Our findings suggest that collapsing glomerulopathy can be a complication of the type I interferonopathy AGS and that a constitutively enhanced type I IFN response in CD133-positive PECs can drive collapsing glomerulopathy.
- Published
- 2021
14. Erratum for the Research Article: 'Acute kidney injury promotes development of papillary renal cell adenoma and carcinoma from renal progenitor cells' by A. J. Peired, G. Antonelli, M. L. Angelotti, M. Allinovi, F. Guzzi, A. Sisti, R. Semeraro, C. Conte, B. Mazzinghi, S. Nardi, M. E. Melica, L. De Chiara, E. Lazzeri, L. Lasagni, T. Lottini, S. Landini, S. Giglio, A. Mari, F. Di Maida, A. Antonelli, F. Porpiglia, R. Schiavina, V. Ficarra, D. Facchiano, M. Gacci, S. Serni, M. Carini, G. J. Netto, R. M. Roperto, A. Magi, C. F. Christiansen, M. Rotondi, H. Liapis, H.-J. Anders, A. Minervini, M. R. Raspollini, P. Romagnani
- Author
-
Marco Allinovi, George Netto, Carolina Conte, Alessandro Antonelli, Giulia Antonelli, and Mario Rotondi
- Subjects
General Medicine - Published
- 2020
- Full Text
- View/download PDF
15. MO060ACUTE KIDNEY INJURY PROMOTES DEVELOPMENT OF A PAPILLARY RENAL CELL ADENOMA-CARCINOMA SEQUENCE FROM RENAL PROGENITORS
- Author
-
Paola Romagnani, Giulia Antonelli, Maria Lucia Angelotti, Francesco Guzzi, Marco Allinovi, and Anna Julie Peired
- Subjects
Transplantation ,Kidney ,Pathology ,medicine.medical_specialty ,Papillary renal cell carcinomas ,Adenoma ,business.industry ,Cancer ,urologic and male genital diseases ,medicine.disease ,female genital diseases and pregnancy complications ,medicine.anatomical_structure ,Nephrology ,Renal cell carcinoma ,medicine ,Carcinoma ,Stem cell ,business ,Mitosis - Abstract
Background and Aims Renal cell carcinoma (RCC) accounts for 2% of all cancers, with about 190,000 new cases per year worldwide. Risk factors for RCC include obesity, diabetes, hypertension and genetic factors, but the majority of cancers occur in apparent absence of clear risk factors. Acute tissue injury (AKI) causes DNA damage and repair processes involving increased cell mitosis and polyploidization, leading to cell function alterations that may potentially drive cancer development. We proposed to verify whether AKI plays a role in RCC development, and to identify the cellular origin of RCC. Method We used the following techniques: 1. observational, retrospective clinical trial to identify a possible association of AKI with RCC. 2. Experimental AKI induction in wild-type mice to study tumor development over 36 weeks. 3. Analysis of TCGA Research Network dataset on human papillary RCC (pRCC) molecular characterization, focusing on AKI-driven pathways. 4. Development of mouse models in which the intracellular domain of Notch 1 (NICD1), a molecule modulated during AKI, is expressed constitutively by all Pax8+ tubular epithelial cells (Pax8/NICD1) or only by Pax2+ renal progenitors (Pax2/NICD1) upon induction in adult mice. The mice were sacrificed at 36 weeks or 4 weeks after AKI. 5. Clonal analysis of tumoral lesions with Confetti reporter. 6. Examination of single cell RNA sequencing (RNAseq) data from pRCC patients. Results We observed that an AKI episode is a major risk factor for pRCC development and recurrence in patients. Wild-type mice subjected to AKI developed pRCC over time in an adenoma-carcinoma sequence, corroborating our human findings. Among AKI-related pathways, Notch1 overexpression in human pRCC associated with worse outcome, prompting us to generate Notch1-overexpressing mice. At 36 weeks o at 4 weeks following AKI, Pax8/NICD1 mice presented a significant decline of renal excretory function as well as type 2 pRCCs. Confetti lineage tracing showed that most of the pRCCs were monoclonal or biclonal, suggesting that they could originate from a local stem cell/progenitor population. Pax2/NICD1 mice presented type 2 pRCCs, and lineage tracing identified single Pax2+ tubular progenitors as the source of pRCCs. Single cell RNAseq analysis confirmed that the molecular signature of the pRCC cell of origin matched the one of human tubular progenitors. Conclusion This study expose the link between AKI and pRCC development in patients, with important clinical implications. In mice, AKI promotes long-term development of type 2 papillary tumors by activating the AKI-associated Notch1 pathway. Additionally, pRCC originates from clonal proliferation of renal progenitors in a classical adenoma-carcinoma sequence leading to invasive pRCC growth and metastatization in mice.
- Published
- 2020
- Full Text
- View/download PDF
16. MO065TUBULAR EPITHELIAL CELL POLYPLOIDIZATION IS REQUIRED TO SURVIVE AKI BUT PROMOTES CKD DEVELOPMENT
- Author
-
Maria Elena Melica, Paola Romagnani, Elena Lazzeri, Giulia Antonelli, Letizia De Chiara, Carolina Conte, Maria Lucia Angelotti, Benedetta Mazzinghi, Laura Lasagni, and Anna Julie Peired
- Subjects
Transplantation ,business.industry ,030232 urology & nephrology ,food and beverages ,030204 cardiovascular system & hematology ,Cell cycle ,medicine.disease ,Epithelium ,World health ,Nuclear translocation ,3. Good health ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,Downregulation and upregulation ,Nephrology ,Fibrosis ,medicine ,Cancer research ,Cell separation ,business ,Protein overexpression - Abstract
Background and Aims Acute kidney injury (AKI) is a global health concern. If not lethal in the acute phase, AKI is considered reversible based on the capacity of surviving tubular cells (TECs) to re-enter cell cycle. However, even mild AKI episodes carry a substantial risk of developing chronic kidney disease (CKD). The pathophysiological basis for this phenomenon remains unclear. Recently, we demonstrated that tubular epithelial cells (TECs) can undergo endoreplication-mediated hypertrophy after AKI. Endoreplications are incomplete cell cycles that lead to the formation of polyploid cells. As polyploid cells can provide increased cell function without restoring tissue integrity, we hypothesized that this mechanism is essential to survive AKI but it can be potentially maladaptive. Method To address this hypothesis, we employed a series of in vitro and in vivo transgenic models based on the Fluorescence Ubiquitin Cell Cycle Indicator (FUCCI) technology to monitor cell cycle phasing in combination with YAP1 overexpression or downregulation. In the in vivo models, YAP1 overexpressing mice and YAP1 knock-out mice were subjected to unilateral ischemia reperfusion injury (IRI) or glycerol-induced rhabdomyolysis to induce AKI. Polyploid cells have been then characterized by microarray analysis, cell sorting, super-resolution STED microscopy and transmission electron microscopy. Results In vitro, human renal tubular cells undergo polyploidization. The fraction of polyploid cells significantly decreases when YAP1 nuclear translocation is blocked, suggesting a possible involvement of YAP1 in regulating TEC polyploidization. After AKI in mice, the inhibition of YAP1 significantly reduces the number of polyploid cells and worsens kidney function resulting in a dramatic decrease of mouse survival. In contrast, YAP1 overexpression leads to an increase in the number of polyploid cells up to 20% of all TECs, further confirming the role of YAP1 in controlling TEC polyploidization. In YAP1 overexpressing mice, electron microscopy and STED analysis revealed the presence of both mononucleated and binucleated polyploid cells. Strikingly, these mice appear to be more prone to develop tubulointerstitial fibrosis acquiring a marked senescent phenotype along with significant decline in renal function thus suggesting an association between polyploidization and CKD development. Indeed, isolation of polyploid cells proved that these cells actively transcribe and secrete pro-fibrotic and senescent factors confirming their role in CKD progression. Conclusion These data suggest that: 1) polyploidization after AKI is required to preserve renal function in the acute phase of damage and it is essential for survival 2) polyploid cells are pro-fibrotic and senescent leading in the long run to the progression of AKI to CKD.
- Published
- 2020
- Full Text
- View/download PDF
17. Erratum to: MO065 tubular epithelial cell polyploidization is required to survive AKI but promotes CKD development
- Author
-
Letizia De Chiara, Elena Lazzeri, Maria Lucia Angelotti, Carolina Conte, Anna Julie Peired, Giulia Antonelli, Maria Elena Melica, Benedetta Mazzinghi, Laura Lasagni, and Paola Romagnani
- Subjects
Transplantation ,Nephrology - Published
- 2022
- Full Text
- View/download PDF
18. CXCL12 blockade preferentially regenerates lost podocytes in cortical nephrons by targeting an intrinsic podocyte-progenitor feedback mechanism
- Author
-
Shrikant R. Mulay, Maria Elena Melica, Duccio Lombardi, Maria Lucia Angelotti, Simone Romoli, Paola Romagnani, Giulia Antonelli, Laura Lasagni, Santhosh Kumar Vr, Carolina Conte, Sven Klussmann, Lidia Anguiano Gómez, Dirk Eulberg, Dana Thomasova, Jyaysi Desai, and Hans-Joachim Anders
- Subjects
Male ,0301 basic medicine ,Kidney Glomerulus ,030232 urology & nephrology ,urologic and male genital diseases ,Kidney ,Podocyte ,Mice ,0302 clinical medicine ,Cells, Cultured ,Feedback, Physiological ,Microscopy, Confocal ,Glomerulosclerosis, Focal Segmental ,Podocytes ,Stem Cells ,Cell Differentiation ,progenitor cells ,Aptamers, Nucleotide ,3. Good health ,Cell biology ,STED ,Treatment Outcome ,medicine.anatomical_structure ,Nephrology ,embryonic structures ,biological phenomena, cell phenomena, and immunity ,Stem cell ,glomerulosclerosis ,Notch signaling pathway ,Mice, Transgenic ,Biology ,Article ,03 medical and health sciences ,Imaging, Three-Dimensional ,medicine ,Animals ,Humans ,Progenitor cell ,FSGS ,proteinuria ,regeneration ,Progenitor ,urogenital system ,Glomerulosclerosis ,Epithelial Cells ,Bowman Capsule ,medicine.disease ,Chemokine CXCL12 ,biological factors ,Blockade ,Disease Models, Animal ,030104 developmental biology ,Doxorubicin ,Glomerular Filtration Barrier - Abstract
Insufficient podocyte regeneration after injury is a central pathomechanism of glomerulosclerosis and chronic kidney disease. Podocytes constitutively secrete the chemokine CXCL12, which is known to regulate homing and activation of stem cells; hence we hypothesized a similar effect of CXCL12 on podocyte progenitors. CXCL12 blockade increased podocyte numbers and attenuated proteinuria in mice with Adriamycin-induced nephropathy. Similar studies in lineage-tracing mice revealed enhanced de novo podocyte formation from parietal epithelial cells in the setting of CXCL12 blockade. Super-resolution microscopy documented full integration of these progenitor-derived podocytes into the glomerular filtration barrier, interdigitating with tertiary foot processes of neighboring podocytes. Quantitative 3D analysis revealed that conventional 2D analysis underestimated the numbers of progenitor-derived podocytes. The 3D analysis also demonstrated differences between juxtamedullary and cortical nephrons in both progenitor endowment and Adriamycin-induced podocyte loss, with more robust podocyte regeneration in cortical nephrons with CXCL12 blockade. Finally, we found that delayed CXCL12 inhibition still had protective effects. In vitro studies found that CXCL12 inhibition uncoupled Notch signaling in podocyte progenitors. These data suggest that CXCL12-driven podocyte-progenitor feedback maintains progenitor quiescence during homeostasis, but also limits their intrinsic capacity to regenerate lost podocytes, especially in cortical nephrons. CXCL12 inhibition could be an innovative therapeutic strategy in glomerular disorders., Graphical abstract
- Published
- 2018
- Full Text
- View/download PDF
19. Endocycle-related tubular cell hypertrophy and progenitor proliferation recover renal function after acute kidney injury
- Author
-
Elisa Ronconi, Sara Nardi, Alexa Burger, Maria Lucia Angelotti, Carolina Conte, Hans-Joachim Anders, Laura Lasagni, Letizia De Chiara, Giulia Antonelli, Paola Romagnani, Alessandro Sisti, Anna Julie Peired, Duccio Lombardi, Francesca Becherucci, Francesco Annunziato, Laura Maggi, Maria Elena Melica, Julian A. Marschner, Ricardo Romero Guevara, Elena Lazzeri, Beat W. Schaefer, Benedetta Mazzinghi, University of Zurich, and Romagnani, Paola
- Subjects
Male ,0301 basic medicine ,Cell division ,030232 urology & nephrology ,General Physics and Astronomy ,urologic and male genital diseases ,Muscle hypertrophy ,Mice ,0302 clinical medicine ,Single-cell analysis ,lcsh:Science ,Multidisciplinary ,Cell Cycle ,Acute kidney injury ,Acute Kidney Injury ,Cell cycle ,3100 General Physics and Astronomy ,Cell biology ,Adult Stem Cells ,Kidney Tubules ,Female ,Single-Cell Analysis ,Science ,Mice, Transgenic ,610 Medicine & health ,1600 General Chemistry ,Cell Enlargement ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,PAX8 Transcription Factor ,03 medical and health sciences ,1300 General Biochemistry, Genetics and Molecular Biology ,medicine ,Animals ,Humans ,Regeneration ,Cell Lineage ,acute kidney injury, renal progenitors, endocycle, stem cell, renal failure ,Progenitor cell ,Progenitor ,Ploidies ,urogenital system ,Regeneration (biology) ,PAX2 Transcription Factor ,Epithelial Cells ,General Chemistry ,Cell Dedifferentiation ,medicine.disease ,Histone Deacetylase Inhibitors ,Mice, Inbred C57BL ,030104 developmental biology ,10036 Medical Clinic ,lcsh:Q - Abstract
Acute kidney injury (AKI) is considered largely reversible based on the capacity of surviving tubular cells to dedifferentiate and replace lost cells via cell division. Here we show by tracking individual tubular cells in conditional Pax8/Confetti mice that kidney function is recovered after AKI despite substantial tubular cell loss. Cell cycle and ploidy analysis upon AKI in conditional Pax8/FUCCI2aR mice and human biopsies identify endocycle-mediated hypertrophy of tubular cells. By contrast, a small subset of Pax2+ tubular progenitors enriches via higher stress resistance and clonal expansion and regenerates necrotic tubule segments, a process that can be enhanced by suitable drugs. Thus, renal functional recovery upon AKI involves remnant tubular cell hypertrophy via endocycle and limited progenitor-driven regeneration that can be pharmacologically enhanced., The recovery of function upon acute kidney injury is thought to involve tubular cell dedifferentiation and proliferation. Here the authors show that Pax2+ progenitors regenerate tubules via cell division while other tubular cells support function recovery by undergoing hypertrophy through endoreplication.
- Published
- 2018
20. SaO032PAPILLARY RENAL CELL CARCINOMA ORIGINATES FROM A POPULATION OF RENAL PROGENITOR CELLS AND IS PROMOTED BY ACUTE KIDNEY INJURY
- Author
-
Maria Lucia Angelotti, Alessandro Sisti, Maria Rosaria Raspollini, Paola Romagnani, Anna Julie Peired, Giulia Antonelli, and Marco Allinovi
- Subjects
Transplantation ,Pathology ,medicine.medical_specialty ,education.field_of_study ,business.industry ,Population ,Acute kidney injury ,medicine.disease ,Nephrology ,Renal cell carcinoma ,medicine ,Progenitor cell ,business ,education - Published
- 2018
- Full Text
- View/download PDF
21. Il documento metodologico
- Author
-
Giulia Antonelli and Fulvio Fabris
- Published
- 2010
- Full Text
- View/download PDF
22. Imaging the kidney: from light to super-resolution microscopy
- Author
-
Giulia Antonelli, Maria Lucia Angelotti, Paola Romagnani, and Carolina Conte
- Subjects
0301 basic medicine ,kidney biopsy ,030232 urology & nephrology ,Reviews ,Kidney ,Mass spectrometry imaging ,law.invention ,03 medical and health sciences ,0302 clinical medicine ,stem cells ,law ,Confocal microscopy ,proximal tubule ,Microscopy ,Image Processing, Computer-Assisted ,Medical imaging ,Animals ,Humans ,Medicine ,AcademicSubjects/MED00340 ,Transplantation ,Super-resolution microscopy ,business.industry ,Cryoelectron Microscopy ,Resolution (electron density) ,Molecular Imaging ,podocytes ,030104 developmental biology ,Microscopy, Fluorescence ,Nephrology ,immunohistochemistry ,Ultrastructure ,Kidney Diseases ,Electron microscope ,business ,Software ,Biomedical engineering - Abstract
The important achievements in kidney physiological and pathophysiological mechanisms can largely be ascribed to progress in the technology of microscopy. Much of what we know about the architecture of the kidney is based on the fundamental descriptions of anatomic microscopists using light microscopy and later by ultrastructural analysis provided by electron microscopy. These two techniques were used for the first classification systems of kidney diseases and for their constant updates. More recently, a series of novel imaging techniques added the analysis in further dimensions of time and space. Confocal microscopy allowed us to sequentially visualize optical sections along the z-axis and the availability of specific analysis software provided a three-dimensional rendering of thicker tissue specimens. Multiphoton microscopy permitted us to simultaneously investigate kidney function and structure in real time. Fluorescence-lifetime imaging microscopy allowed to study the spatial distribution of metabolites. Super-resolution microscopy increased sensitivity and resolution up to nanoscale levels. With cryo-electron microscopy, researchers could visualize the individual biomolecules at atomic levels directly in the tissues and understand their interaction at subcellular levels. Finally, matrix-assisted laser desorption/ionization imaging mass spectrometry permitted the measuring of hundreds of different molecules at the same time on tissue sections at high resolution. This review provides an overview of available kidney imaging strategies, with a focus on the possible impact of the most recent technical improvements.
- Full Text
- View/download PDF
23. Acute kidney injury promotes development of papillary renal cell adenoma and carcinoma from renal progenitor cells
- Author
-
Hans-Joachim Anders, Alessandro Antonelli, Tiziano Lottini, Andrea Minervini, Marco Carini, Francesco Guzzi, Paola Romagnani, Mauro Gacci, Alessandro Sisti, Carolina Conte, Fabrizio Di Maida, Mario Rotondi, Samuela Landini, Marco Allinovi, Maria Elena Melica, Rosa Maria Roperto, Francesco Porpiglia, Vincenzo Ficarra, Benedetta Mazzinghi, Sergio Serni, Sabrina Giglio, George J. Netto, Maria Rosaria Raspollini, Maria Lucia Angelotti, Letizia De Chiara, Sara Nardi, Giulia Antonelli, Davide Facchiano, Alberto Magi, Riccardo Schiavina, Elena Lazzeri, Helen Liapis, Andrea Mari, Roberto Semeraro, Christian Fynbo Christiansen, Laura Lasagni, Anna Julie Peired, Peired A.J., Antonelli G., Angelotti M.L., Allinovi M., Guzzi F., Sisti A., Semeraro R., Conte C., Mazzinghi B., Nardi S., Melica M.E., De Chiara L., Lazzeri E., Lasagni L., Lottini T., Landini S., Giglio S., Mari A., Di Maida F., Antonelli A., Porpiglia F., Schiavina R., Ficarra V., Facchiano D., Gacci M., Serni S., Carini M., Netto G.J., Roperto R.M., Magi A., Christiansen C.F., Rotondi M., Liapis H., Anders H.-J., Minervini A., Raspollini M.R., and Romagnani P.
- Subjects
Adenoma ,0301 basic medicine ,papillary renal cell carcinoma ,medicine.disease_cause ,urologic and male genital diseases ,DISEASE ,PATHWAY ,PROTECTS ,Mice ,03 medical and health sciences ,0302 clinical medicine ,AKI ,NOTCH1 ,REGENERATION ,Renal cell carcinoma ,Biomarkers, Tumor ,ABLATION ,medicine ,Carcinoma ,Animals ,Progenitor cell ,RECURRENCE ,Carcinoma, Renal Cell ,RECEPTOR ,Papillary renal cell carcinomas ,business.industry ,Stem Cells ,Papillary Adenoma ,Acute kidney injury ,renal carcinoma ,General Medicine ,Acute Kidney Injury ,medicine.disease ,TUMORS ,Kidney Neoplasms ,3. Good health ,NOTCH ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cancer research ,GROWTH ,Neoplasm Recurrence, Local ,Stem cell ,Carcinogenesis ,business - Abstract
Acute tissue injury causes DNA damage and repair processes involving increased cell mitosis and polyploidization, leading to cell function alterations that may potentially drive cancer development. Here, we show that acute kidney injury (AKI) increased the risk for papillary renal cell carcinoma (pRCC) development and tumor relapse in humans as confirmed by data collected from several single-center and multicentric studies. Lineage tracing of tubular epithelial cells (TECs) after AKI induction and long-term follow-up in mice showed time-dependent onset of clonal papillary tumors in an adenoma-carcinoma sequence. Among AKI-related pathways, NOTCH1 overexpression in human pRCC associated with worse outcome and was specific for type 2 pRCC. Mice overexpressing NOTCH1 in TECs developed papillary adenomas and type 2 pRCCs, and AKI accelerated this process. Lineage tracing in mice identified single renal progenitors as the cell of origin of papillary tumors. Single-cell RNA sequencing showed that human renal progenitor transcriptome showed similarities to PT1, the putative cell of origin of human pRCC. Furthermore, NOTCH1 overexpression in cultured human renal progenitor cells induced tumor-like 3D growth. Thus, AKI can drive tumorigenesis from local tissue progenitor cells. In particular, we find that AKI promotes the development of pRCC from single progenitors through a classical adenoma-carcinoma sequence.
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.