Skip to main content
American Journal of Clinical and Experimental Urology logoLink to American Journal of Clinical and Experimental Urology
. 2022 Dec 25;10(6):449–461.

Current research and future directions in non-malignant urologic research - proceedings of the annual CAIRIBU meeting

Petra Popovics 1, Kristina L Penniston 2
PMCID: PMC9831912  PMID: 36636691

Abstract

The Annual Collaborating for the Advancement of Interdisciplinary Research (CAIRIBU) Meeting in 2022 highlighted basic, translational, and clinical non-malignant urology research within five main areas affecting the urinary tract: urinary dysfunction due to prostate disease, microbes and infection, bladder function and physiology, neurology and neuromuscular influences and calculi and obstruction. In this paper, we summarize main findings and future directions outlined by CAIRIBU-affiliated scientists who presented as part of the scientific sessions.

Keywords: Benign urologic research, prostate, bladder, urogenital tract

Introduction

The annual meeting of the National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK)-funded group of U54 Urology O’Brien Centers, P20 Urology Centers, and K12 Urology Career Development Programs took place in December 2022. Known as CAIRIBU - Collaborating for the Advancement of Interdisciplinary Research in Benign Urology - the annual meeting is one of the primary mechanisms for exchanging knowledge about the non-malignant genitourinary research in which CAIRIBU investigators are involved. CAIRIBU was formed in 2018 by Program Officers with the NIDDK (see accompanying commentary paper in this issue for further details). A primary objective was to stimulate interactions between investigators leading to cross-disciplinary research that may be better able to address current knowledge gaps in the function and physiology of the genitourinary system and lower urinary tract. The meeting is organized each year by the CAIRIBU (U24) Interactions Core (PI, Kristina Penniston, PhD) in the Department of Urology at the University of Wisconsin School of Medicine and Public Health. This meeting brief provides a summary of the science presented by CAIRIBU-affiliated trainees and investigators at the 2022 CAIRIBU meeting.

Lower urinary tract symptoms/dysfunction (LUTS/D) and prostate

LUTS/D as a consequence of aging

LUTS/D related to non-malignant pathological changes in the prostate, also known as Benign Prostatic Hyperplasia (BPH), substantially deteriorate the quality of life of aging men. Most bothersome symptoms include nocturia, urgency and incomplete emptying, but BPH often leads to acute urinary retention with the inability to urinate and the need for long-term catheterization [1]. Age is the single most important risk factor in LUTS/D which was the focus of the research presented by Scott Bauer MD, ScM, Assistant Professor and former K12 Scholar at University of California San Francisco. Mitochondrial dysfunction has been associated with aging; patients with inherited mitochondrial diseases have significantly higher rate of urinary urgency, weak stream, incontinence and incomplete emptying [2,3]. Dr. Bauer’s research aims to elucidate whether mitochondrial content, protein expression and bioenergetic profile differ in the prostates of BPH patients and healthy individuals. He is also developing a prospective cohort of men and women to assess the association of symptom development and muscle function.

The role of resident and infiltrating cells in prostate disease

It is currently accepted that the prostate undergoes multiple pathological changes, including growth of epithelial and stromal nodules, smooth muscle dysfunction, inflammation and fibrosis [4,5]. Research has been traditionally focusing on enlargement and smooth muscle physiology, whereas inflammation and fibrosis are understudied and pharmacologically untargeted. Fibrosis is the result of a developing imbalance between extracellular matrix (ECM) deposition and degradation with the former becoming progressively dominant over time [6]. Chronic inflammation has been recognized as the main driver of fibrosis in multiple diseases [7]. The dominant immune cell types in the prostate are T-cells and macrophages [8] and the mechanism attracting these cells to the prostate and their role in disease pathogenesis are important questions motivating the current goals of the non-malignant prostate research community.

Via sampling epithelial and stromal BPH nodules as well as normal prostate areas from the peripheral zone, the team of Jonathan Pollack MD, PhD at the Stanford University U54 Urology O’Brien Center (PI, James Brooks, MD) identified that chemokine (C-X-C motif) ligand 13 (CXCL13) expression was highly upregulated in BPH fibroblasts. CXCL13 expression was also associated with T- and B-cell infiltration [9]. Their future work will focus on B- and T-cell receptor repertoire to aid in identifying specific adaptive immune responses as well as to characterize T-cell subsets and the molecular drivers of CXCL13 expression.

The research aims of Petra Popovics, PhD (Assistant Professor at Eastern Virginia Medical School and formerly a K12 Scholar at UW-Madison) are to identify the prostate immune landscape that develops in response to steroid hormone imbalance in a mouse model replicating hormone levels in the aging human prostate [10]. Interestingly, she observed an increase in macrophages but not in T-cells, as well as the migration of macrophages to the prostate lumen differentiating into lipid-accumulating cells aka “foam cells”. Foam cells were also observed in human BPH tissues in association with regional lipid accumulation sites. Future goals of the Popovics Lab will be to identify what cell types are the main sources of foam cells, what mechanisms regulate macrophage migration to the luminal space and which factors drive lipid accumulation in LUTS/D.

Studying the hallmarks of prostatic fibrosis in the human prostate and identifying which prostatic cell type is responsible for this pathological change have been the main goals of Chad Vezina, PhD (Co-Director of the UW-Madison U54 Urology O’Brien Center). His laboratory identified that collagen density in the prostate increases near the urethra only in BPH patients where it is most likely to drive urinary symptoms. Using lineage tracing with the Cre/lox technology, his team also showed that myeloid cells and fibroblasts are the major source of collagen deposition. Dr. Vezina’s future work will focus on identifying molecules that attract pro-fibrotic myeloid cells to the prostate.

The work of Jill Macoska, PhD (Professor at the University of Massachusetts Boston and Project Leader in the UW-Madison U54 Urology O’Brien Center) has been focusing primarily on how fibroblasts become ECM-producing factories and promote prostatic fibrosis. By investigating cytokines that are upregulated in prostate fibroblasts in aging men, her group at the University of Massachusetts has identified a significant increase in CXCL12 expression [11] which drives conversion of fibroblasts to myofibroblasts, a transition that results in increased production of collagen I [12,13]. Moreover, the use of an antagonist of the CXCL12 receptor (CXCR4) ameliorated high fat diet-induced periurethral collagen accumulation and urinary dysfunction [14]. In contrast, her most recent studies have shown that the interleukin (IL) 4/IL13/signal transducer and activator of transcription 6 (STAT6) axis is also pro-inflammatory in the prostate, but it does not transform fibroblasts to myofibroblasts. Rather, the stimulation of the pathway generates activated fibroblasts that preserve their proliferative potential [15]. Future directions of the Lab include studying fibroblast chemotaxis and characterization of fibroblast plasticity and resistance to apoptosis in response to IL4 and IL13.

Specimen and data resources are spare in non-malignant prostate research in part due to surgeries not prioritizing the preservation of tissue integrity and the lack of normal “healthy” tissue that could be utilized as controls. Douglas Strand, PhD, Associate Professor at the University of Texas Southwestern Medical Center and Project Leader in the UW-Madison U54 O’Brien Center has developed a prostate biorepository utilizing prostatectomy specimens and normal prostates from organ donors with associated patient data that now fuels his and his collaborators’ research. In addition, using single-cell RNA sequencing (scRNAseq), his lab identified the large diversity across epithelial (basal, luminal, club, hillock cells) and stromal cells (peri-epithelial and interstitial) in the prostate [16-18]. Future directions in his lab include providing a comprehensive hierarchical definition of all human prostate cells, a human bladder cell reference atlas as well as to create a multi-omic analysis of the lower urinary tract.

Imaging fibrosis in the prostate

As the pathological diversity of BPH is becoming more and more apparent, the clinical implementation of novel diagnostic strategies identifying pathological features has become a crucial goal in non-malignant urology research. Shane Wells, MD, an Opportunity Pool Award recipient from the UW-Madison U54 Urology O’Brien Center, tackles how gross pathological abnormalities in the prostate can be visualized by imaging techniques. He uses elastography, which is an image-based, quantitative form of palpitation involving low-frequency sounds waves transmitted into the tissue followed by their detection. The stiffness map presented by Dr. Wells outlined how increased stiffness in the prostate transitional zone can be visualized. He also identified that the thickness of the fibrotic capsule increases with prostate volume. Dr. Wells’ collaboration with Alejandro Roldán-Alzate, PhD (also of the UW-Madison U54 Urology O’Brien Center) aims to characterize bladder emptying during voiding using dynamic MRI. Preliminary results are pointing to a urethral opening dysfunction in BPH patients.

Microbes and infection in the genitourinary tract

Urobiome composition in children in health and disease

Recent innovations in collection techniques and the launch of next generation sequencing for the identification of microbial strains have allowed the detection of uncultured microbial strains and led to the confirmation of a urobiome in 2014 [19-21]. Interestingly, about 60% of urogenital microbiome strains are shared with the gut and 30% with the vagina in women while various unique elements are found only in the urobiome [22]. Although increasingly more data are becoming available deciphering the adult urobiome, research in children is far less developed. Characterization of the healthy urobiome in infants is the goal of Seth Reasoner, a graduate student at Vanderbilt University in the laboratory of Maria Hadjifrangiskou, (PI of the Vanderbilt P20 Exploratory Center). The team has developed a collection strategy by targeting infant males (4-6 month) undergoing circumcision and catheterization. They combined blood or brucella agar urine culturing with MALDI-TOF-based identification and 16S rRNA sequencing. Importantly, they included saline flushed through the catheter as a control. This strategy has identified Actinotignum schaalii as a predominant strain in the healthy infant urinary tract. Going forward, the group is preparing to make their investigation publicly available and searchable and hope to utilize their standardized method to track age-related changes in urobiome composition. Maryellen Kelly, DNP, MHSc, a former K12 Scholar from Duke University, assessed urobiome composition in children up to five years of age with sequencing achieving species-specific resolution. Her team concentrated samples with vacuum filtration to overcome low biomass in the samples. They found that diversity was associated with age and with female sex and identified the predominance of Prevotella, Schaalia and Fusobacterium genera. Their future goal is to investigate the stability of the pediatric urobiome over time and compare the microbiomes of children with or without recurrent urinary tract infection (UTI). A more disease-focused approach was utilized by Miguel Verbitsky, PhD, from the lab of Ali Gharavi, MD (one of the PIs of the Columbia University U54 Urology O’Brien Center), who compared children with or without vesicoureteral reflux (VUR) who had UTIs. Using 16S rRNA sequencing, they identified that urobiota diversity was higher in children who had UTI but no VUR and that microbial diversity correlated with age. They performed a metabolite genome-wide association study (mGWAS) to investigate the relationship between genetic factors and the urine metabolome. This analysis highlighted potential involvement of host genes associated with immune surveillance, inflammation and genitourinary tract development and diseases with the abundance of certain bacterial taxa.

Mechanism of colonization during urinary tract infections UTI

Although our knowledge on the existence of uropathogenic microbes preceded the discovery of the urobiome, research still falls short of eradicating these infections. UTI is diagnosed in 150 million people worldwide and in 7 million in the U.S. annually, whereas 15% of all antibiotic prescriptions are prescribed for UTI treatment. The majority of UTI cases involve uropathogenic E. coli (UPEC) strains [23]. Maria Hadjifrangiskou, PhD at Vanderbilt University (P20 Center PI) focuses on how E. coli is internalized in urothelial cells, how it develops intracellular biofilms/populations and what stress this imposes on the host cell. She discovered that in the E. coli biofilm, bacterial proteins, including quinol oxidase complexes, are spatially organized and coordinated by oxygen gradient [24]. The team identified that bacteria utilize the host cell’s oxygen which leads to increased HIF-1α level thereby suppressing apoptosis and shifting the cells to glycolysis. Dr. Hadjifrangiskou’s future studies will focus on understanding the role of HIF-1α in the progression of UTI. Tatyana Sysoeva, PhD, an Assistant Professor at the University of Alabama in Huntsville (and a former Duke University K12 Scholar) is studying horizontal gene transfer and multidrug resistance genes carried on the bacterial F-plasmid [25]. Her team showed that the bivalent action of the TraT factor prevents conjugation-related plasmid transfer, but also protects from phages, some antibiotics, detergents and serum complement. Her lab also discovered that TraT forms oligomers and interacts with bacterial outer membrane proteins. The team’s future studies will decipher the role of TraT in UPEC serum resistance.

During a UTI, it is vital for the bacteria to capture iron from the host, which is needed for DNA replication and metabolism. Sources are elemental iron that are taken up by siderophores or via the recovery of iron from heme, transferrin or lactoferrin from the environment [26]. Jonathan Barasch, MD, PhD, one of the PIs of the Columbia University U54 Urology O’Brien Center, studies bacterial iron uptake during UTI. He has identified that bacteria cause damage in the epithelium which releases iron that in turn acts as a bacterial growth factor. Dr. Barasch has also discovered that the urothelium expresses NGAL upon bacterial infection, which blocks the transit of iron to bacteria by inhibiting bacterial siderophores [27]. Hematuria is also a hallmark of UTI which activates Heme-associated proteins and a defense mechanism against iron in the urothelium. The heme transporter, Slc48a1, is induced in the urothelium in a temporally different manner in female and male mice whereas HMOX1 splits heme intracellularly. Knocking out the heme transporter is protective in UTI, according to Dr. Barasch, due to the toxic effect of iron-induced ferroptosis, necroptosis and apoptosis.

Macrophage diversity in the bladder

Describing the normal macrophage populations and their change during the UTI immune response has been the focus of Nicholas Steers, PhD, an Opportunity Pool Award recipient from the Columbia University U54 Urology O’Brien Center. For a long time, it has been accepted that tissue resident macrophages are self-maintained and independent of hematopoietic cells, however, this dogma has shifted most recently [28,29]. Hematopoietic stem cells/monocytes arrive from the yolk sac, fetal liver and bone marrow, depending on the stage of development, to compose and replenish the tissue resident macrophage population [30], however, this process has been understudied in the bladder. Using a deep phenotyping flow cytometry panel for macrophages, Dr. Steers identified five subpopulations of macrophages. More interestingly, he detected substantial changes in two subpopulations in response to UTI. His future goals include identifying the origin of macrophages via lineage tracing, studying the proliferative potential of tissue resident macrophages and their replenishment from monocytes.

Bladder function and physiology

Mechanical modeling of the bladder

The urinary bladder’s simple mechanistic role is to store and empty urine. These functions are coupled to a highly complex tissue architecture involving a longitudinal and a circular smooth muscle layer (detrusor), a mucosa and a specialized epithelium known as the urothelium. Mechanical properties of bladder filling have been the major goal of Eli Broemer, a graduate student in the Lab of Sara Roccabianca, PhD at Michigan State University. He used an imaging technique developed in the laboratory of Nathan Tykocki, PhD (also at Michigan State University), which registers bladder filling simultaneously using mirrors from different directions, and extracted a 3D model using spatial carving. From this, he modeled the volumetric change over time and associated it with the change in pressure. Comparing male and female mouse bladders, the study determined that bladder wall stress increases rapidly when the bladder reaches capacity and that males had higher stress in the bladder wall than females. Also, he identified that the bladder stretches more in the circumferential than the longitudinal direction i.e. possesses an anisotropic behavior.

Urothelial changes in injury, diabetes and aging

The urothelium exhibits a crucial role in allowing repeated stretching of the bladder during filling, but also provides an important barrier function against pathogenic microbes, toxins and carcinogens [31,32]. Superficial/umbrella cells have a long life-span, exist as polyploid cells and are replenished from basal cells [33]. The research of Cathy Mendelsohn, PhD (one of the PIs of the Columbia University U54 Urology O’Brien Center) showed that basal cells can undergo transformation/activation during exposure to chemical or infectious agents and acquire squamous markers, leading to the loss of superficial cells. This process can deteriorate the permeability barrier and predispose to carcinogenesis. The Mendelsohn Lab also tackles the role of Pparg in urothelial cell differentiation; Pparg differentiates basal cells to superficial cells, whereas Pparg deletion results in squamous metaplasia [34]. They revealed that Pparg is required to shut down NFκB signaling its deletion resulting in continuous immune cell infiltration. Interestingly, deletion of Pparg in superficial cells makes neutral lipids become trapped in the mitochondria intermembrane space due to defective fatty acid transport [34]. Future studies of the lab will assess the potential beneficial activities of Pparg agonists in repairing the urothelium after recurrent UTI. Apart from external stimuli, aging is an important internal factor that can lead to the deterioration of bladder tissue structure and function [35]. Indira Mysorekar, PhD at the Baylor College of Medicine studies the hallmarks of aging in the urothelium. Her earlier work identified an increase in tertiary lymphoid structures [36]. More recently, using transmission electronmicroscopy, her team showed the accumulation of lysosomes, mitochondria with reduced activity, a decline in autophagy, elevated senescence and cell damage pathways, DNA damage and increased cell cycle arrest in the aged mouse bladder. The aged bladder had constitutively high level of reactive oxygen species (ROS) and deficient antioxidant response and was more susceptible to intracellular biofilm formation of UPEC. The future work of the lab focuses on testing the beneficial effects of D-mannose on reducing the observed age-associated changes in the urothelium. Bladder dysfunction is a common complication in diabetes (diabetic cystopathy) that may persist even with optimal glucose management; therapeutic options are sparse [37]. Michael Odom, PhD, works as a K12 Scholar in the Purves Lab at Duke University. He focuses on detrusor underactivity in diabetes. It is believed that diabetic metabolites activate NLRP3 inflammasome-related pyroptosis in the urothelium providing direct contact of urine with the lamina propria and the smooth muscle layers [38,39]. Using the Akita diabetes mouse model, the team showed that NLRP3 is required for the development of increased voiding volume and decreased frequency associated with the underactive voiding phenotype [40]. Dr. Odom’s current work focuses on testing the utilization of the Prostaglandin F2α receptor (FP) in increasing the contractility of underactive detrusor, and his future work will focus on studying FP receptor populations and assess FP receptor smooth muscle signaling.

Genetic studies in non-malignant urology

Urological diseases, including bladder dysfunction, may have underlying association with an individual’s genetic composition. According to Ali Gharavi, MD, one of the PIs of the Columbia University U54 Urology O’Brien Center, genetic variance may affect 5-10% of patients with congenital urinary tract disorders. Congenital defects associated with genetic alterations that were identified by the group include vesicoureteral reflux, duplicated ureter, multicystic dysplastic kidneys and unilateral renal hypoplasia and can be linked to microdeletion syndromes or copy number variations [35,41,42]. Interestingly, the examination of common alleles in vesicoureteral reflux identified the involvement of developmental genes (eg. WNT5a, BMP5) whereas defective bladder and ureter development was observed in WNT5a KO mice further confirming the association [41]. Going forward, the utilization of available biorepositories for genomics research, including the All of Us Research Hub that includes many benign urological conditions such as urinary incontinence, retention and UTI, will accelerate discoveries in genomics research of other benign urological diseases, including the bladder.

Utilizing machine learning in cystoscopy analysis

Bladder trabeculation has been recognized as a surrogate marker for bladder function which correlates with urodynamic findings [43]. Ramy Goueli, MD, and his team at UT-Southwestern aim to develop a machine learning strategy using convoluted neural network, which is a class of deep neural networks most commonly used to analyze visual imagery. Dr. Goueli’s team performed a retrospective chart review on non-neurogenic male urology patients presenting with LUTS. Cystoscopy videos were graded by two urologists for trabeculation and still images were used to create an algorithm for the presence and grade (none, mild, moderate, severe) of bladder trabeculations [44]. Studying only 27 patients, they acquired 76% accuracy for determining trabeculation grade with this technique. Due to the heterogeneity of bladder trabeculations within individual patients, the methodology was changed to patch-based labeling. This method is currently in testing to improve the detection of severe trabeculations. When the group develops satisfactory automated detection and grading of bladder trabeculations they will use the method to correlate grades with clinical measures of bladder function and also employ similar techniques in the neurogenic bladder population.

Neurology and neuromuscular influences on the urinary tract

Mast cells in neurogenic bladder

Neurogenic bladder can develop when there is a disruption between the CNS and the somatic nervous system [45]. Transient contractions and the rate of pressure increase in the bladder affect sensory outflow and contribute to how bladder fullness is sensed [46]. Organization and composition of the ECM also influences the rate of smooth muscle contractility and drives sensation. Pragya Saxena, a graduate student from the Tykocki Lab at Michigan State University, tackles the role of mast cells in smooth muscle contractility in the bladder based on previous studies showing increased mast cell numbers in interstitial cystitis [47]. Using the Pentaplanar Reflected Image Macroscopy System, she showed that the mast cell activator compound 48/80 increased the amplitude and the slope of contractions. This effect was inhibited by a cyclooxygenase inhibitor that decreases prostaglandin levels. Her future research will identify the re-arrangement of the ECM in the bladder after mast cell activation, and identify prostaglandins that play key roles in this model. B. Malique Jones, also a graduate student from the Tykocki lab, studies how inflammatory factors can induce detrusor overactivity, mimicking neurogenic bladder, independent of afferent and efferent nerve signaling. She determined that the presence of urothelium in the tissue bath is required for compound 48/80 to increase contractility and phasic activity in smooth muscle and to augment nerve-evoked (electrical field stimulation) contractions. Using specific inhibitors, she determined that this was due to ATP and prostaglandins released from the compound 48/80-treated urothelium.

Neuron cross-talk in pain sensation

The work of LaTasha Crawford, VMD, PhD, DACVP (K12 Scholar at UW-Madison) targeted two questions: what if sensory neurons from the bladder can communicate with sensory neurons from the skin? And can these neuron groups cause hyperactivity of others in the same or nearby ganglia? She studies changes in the dorsal root ganglia (DRG) where diverse sensory neurons from different organs may interact [48]. Interestingly, she showed that the sural dermatoma of the foot becomes hypersensitive in the intravesicular acrolein model of cystitis, implying a neuronal cross-talk in DRG. Moreover, while voiding spot frequency resolved, allodynia persisted even after 8 days. The group has also developed an ex vivo skin-ganglion-cord preparation for neurophyisiology assays, an image deconvolution and digital clearing method to visualize DRG neurons and their calcium signaling, whole mount staining of the bladder and used genetic strategies to label afferent neurons in mice. The Crawford Lab is also working on building a comparative neuro biobank of dogs, cats, and horses with dorsal root ganglia preparations and innervated tissues.

Environmental toxicants in bladder function

Kimberly Keil Stietz, PhD, an Assistant Professor at the UW-Madison and Project Leader for the U54 Urology O’Brien Center studies the role of polychlorinated biphenyls (PCBs), manmade compounds that were previously produced for chemical stabilizers, heat dissipaters and paints but which now occur as biproducts of paint production. PCBs affect dendrite morphology, elevate calcium signaling in the brain and cause inflammation [49,50]. Dr. Keil Stietz’s team exposed mice to a mixture of PCBs starting with the mother prior to mating and terminated the exposure at the time of weaning. They observed increased voiding frequency in both females and males, decreased intervals between voids in females and increased maximal bladder pressure in males. She concluded that PCB-enhanced bladder contractility in response to cholinergic stimuli may be partly due to increasing bladder nerve density [51]. Future studies in her lab and in her research related to the Opportunity Pool Award she received from the Stanford University U54 Urology O’Brien Center will assess how sensory function is altered, how inflammation is involved (increase in mast cells, macrophages), and how PCBs change neural activity within the Pontine micturition center.

Physical training for urinary incontinence (UI)

General factors contributing to UI include obesity, autonomic instability, bladder muscle over- or underactivity and pelvic floor weakness. With the onset of aging, additional factors, such as physical function and cognitive decline as well as frailty will also provoke UI events [52,53]. Allison Huang, MD, (co-PI of the UCSF-Kaiser Permanente K12 Urologic Epidemiology Institutional Career Development Program) is interested in whether behavioral interventions that improve overall skeletal muscle, autonomic and general physiological function can prevent or reverse UI. Yoga is an ideal candidate for the development of such intervention because it may incorporate mindful awareness and improved control of pelvic floor muscles as well as improved overall physical conditioning. Moreover, breathing exercises increase peripheral balance and decrease detrusor muscle instability. The team hypothesized that the practice of yoga would lead to an overall increase in continence-related functioning and the improvement of quality of life. To test this hypothesis, Dr. Huang’s team developed a multicenter randomized trial to implement a therapeutic yoga program for UI in older women, the “Lessening Incontinence with Low-Impact Activity (LILA)” trial. It identified that the specialized yoga training significantly decreased UI events more than a regular physical conditioning program in just three months. Future studies of the team will investigate factors that mediate these beneficial effects, explore differences across age groups and in-person vs. virtual training as well as monitor the persistence of improvements.

Calculi and obstruction in the urinary tract

Improving urolithiasis diagnostics with machine learning

The Center for Machine Learning in Urology is a CAIRIBU P20 Exploratory Center (PI, Greg Tasian, MD, MSc, MSCE, University of Pennsylvania Perelman School of Medicine and Children’s Hospital of Philadelphia [CHOP]). The Center has been exploring machine learning techniques to predict urologic outcomes and to aid diagnosis [54-57]. In the first of two talks from the Center, Yuemeng Li, a graduate student in the Center for Biomedical Image Computing and Analytics at the Perelman School of Medicine, described the development of a transformer model of deep learning to automatically identify and measure kidney stones on CT scans, the result of which would mean more efficient, accurate, less burdensome, and standardized quantification of stones within the urinary tract. The first task was image segmentation for which urologists manually reviewed >100 CT urograms from patients of different sizes and ages and manually annotated areas of interest. Machine learning experts then designed a predictive stone mapping model based on an atlas derived from the annotated dataset. After the model achieved acceptable performance (Mean Dice Score 0.96), investigators began training the model to generate spatial maps of kidney stones from non-contrast CT scans of >100 unique patients [58]. Ultimately, the automated stone identification algorithm detected individual kidney stones with 100% sensitivity; spatial mapping (3D reconstruction) efforts to quantify the frequency of stones across subjects are underway.

Building on this work, Perelman School of Medicine medical student Abhay Singh described a machine learning project aimed at predicting ureteral stone passage, the result of which could mean earlier identification of patients likely to successfully pass their stones vs. requiring surgical intervention. A retrospective cohort study was assembled from >250 children and adults who presented to CHOP and University of Pennsylvania with ureteral stones, 48% of whom spontaneously passed their stones (44 and 54% of adults and children, respectively). Axial, vertical, and horizontal stone lengths were available from CTs. Other variables - BMI, hematuria, nausea, fever, flank pain, stone location, and others - were assessed. Random forest modeling, a type of supervised machine learning algorithm, was employed in an attempt to predict stone passage. The adult and pediatric models achieved 63% and 70% accuracy, respectively. Patient age and the orthogonal measurement product (the product of axial vertical and horizontal stone length) were the most predictive variables in both.

Mechanisms and safety of laser ablation in lithotripsy

Two investigators from the Center for Urological Laser Technologies at Duke University (a new CAIRIBU FORWARD Urology P20 Center; PI, Pei Zhong, PhD) delivered presentations related to laser lithotripsy (LL) in surgical stone removal. Ureteroscopic LL using a holmium: YAG laser (YAG = yttrium-aluminum-garnet) is a common procedure for vaporizing ureteral and renal calculi. Fragments may then be either removed with a basket or, if small enough, excreted painlessly by patients after the intervention. A deeper understanding of the mechanisms of LL will inform the development of new and more effective technology. John Dolbow, PhD, Professor in the Department of Mechanical Engineering and Materials Science at Duke University, described efforts in collaboration with investigators from Virginia Tech to understand the relative importance of the photothermal effects vs. fluid-stone interactions of LL. During lithotripsy there is usually continuous irrigation. In this milieu, the holmium: YAG laser pulse generates an elongated cavitation bubble (a type of fluid-stone interaction) that generates a weak shock wave. While conventional wisdom is that cavitation is a primary mechanism for inducing damage to the stone [59], Dolbow described bench top experiments and mathematical modeling that demonstrate an effect of thermal ablation. Because the kidneys are susceptible to thermal injury during LL, his work supports concern about the energy released by the lasers as well as the frequency of pulses delivered [60].

Michael Lipkin, MD described a new type of laser being used during ureteroscopic LL that vaporizes stones to dust, the thulium laser [61]. Stone dusting takes advantage of the much higher pulse frequency that is achievable with new lasers [62], especially the thulium. However, its mechanism of stone ablation is thought to differ from the traditional holmium: YAG laser. Because it is relatively new, the optimal thulium fiber settings for energy and frequency during stone dusting - i.e., those that appropriately balance efficiency and safety - have not yet been identified [63]. Lipkin presented data from porcine studies in which macroscopic tissue damage was observed in kidneys as a result of the heat from thulium laser energy [64]. With this knowledge, the group conducted bench top studies on both artificial and human stones. Results thus far suggest that, as compared to the holmium: YAG laser, the thulium laser should be set on higher energy but lower pulse frequency in order to avoid thermal-related injury. Questions remain about how close the laser tip should be to the stone during LL; this and other knowledge gaps will be addressed by the Duke P20 Center.

Oxalate metabolism in calcium oxalate urolithiasis

Kidney stones affect 1 in 11 Americans; there is increasing prevalence among women [65]. The most common type of kidney stone has some component of calcium oxalate. While multifactorial in etiology, urinary oxalate excretion is a primary risk factor. Urine oxalate represents the sum total of exogenous oxalate (from the diet) as well as endogenous (i.e., that which is synthesized in vivo from precursors in both enzymatic and non-enzymatic pathways) [66]. Sonia Fargue, PhD, from the P20 Exploratory Center at the University of Alabama at Birmingham (PI, Dean Assimos, MD), said that endogenous oxalate pathways, depending on the individual, may account for a significant amount of urine oxalate [67]. She presented results from a clinical study in which intravenous infusions of carbon13 oxalate were administered to both stone formers and non-stone formers in order to measure oxalate synthesis. Urinary oxalate excretion was then examined. Compared to non-stone formers, stone formers with higher body mass excreted more oxalate in urine, even while on diets controlled for oxalate. Interestingly, lean mass but not fat mass was most associated with oxaluria in both patients and controls. Fargue thus suggested a larger than previously-thought role for muscle and bone (both are components of lean body mass) in oxalate synthesis. Weight-bearing muscles and bone are both rich in collagen. Fargue explained that the hydroxyproline produced by post-translational hydroxylation of proline residues in collagen is a glyoxylate precursor [68]. Glyoxylate is then converted to oxalate in a pathway catalyzed by lactate dehydrogenase. These data support the idea that endogenous oxalate synthesis has been previously underestimated and may account for a significant amount of the variability in the urinary oxalate excretion of patients who form calcium oxalate stones [67].

Conclusion

This year, CAIRIBU investigators reported progress in five major areas of non-malignant urologic research summarized in Figure 1. Recent clinical advances included the use of improved techniques in imaging, surgical management and implementing machine learning in analysis. Basic and translational tools have allowed the expansion of our knowledge on cellular composition and behavior as well as disease-driven molecular signaling in the lower urinary tract mainly in the aspects of inflammation, fibrosis and aging. CAIRIBU investigators are collaborating to provide resources (biospecimens, expressional and genetic databases, microbiome composition and cellular atlases) for the larger urology research community.

Figure 1.

Figure 1

Main research areas discussed at the 2022 CAIRIBU Annual Meeting.

Acknowledgements

National Institute of Diabetes and Digestive and Kidney Diseases U24 Interactions Core grant (U24-DK-127726).

Disclosure of conflict of interest

None.

References

  • 1.McVary KT. BPH: epidemiology and comorbidities. Am J Manag Care. 2006;12(Suppl):S122–128. [PubMed] [Google Scholar]
  • 2.Poole OV, Uchiyama T, Skorupinska I, Skorupinska M, Germain L, Kozyra D, Holmes S, James N, Bugiardini E, Woodward C, Quinlivan R, Emmanuel A, Hanna MG, Panicker JN, Pitceathly RDS. Urogenital symptoms in mitochondrial disease: overlooked and undertreated. Eur J Neurol. 2019;26:1111–1120. doi: 10.1111/ene.13952. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Parsons T, Weimer L, Engelstad K, Linker A, Battista V, Wei Y, Hirano M, Dimauro S, De Vivo DC, Kaufmann P. Autonomic symptoms in carriers of the m. 3243A>G mitochondrial DNA mutation. Arch Neurol. 2010;67:976–979. doi: 10.1001/archneurol.2010.174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Rodriguez-Nieves JA, Macoska JA. Prostatic fibrosis, lower urinary tract symptoms, and BPH. Nat Rev Urol. 2013;10:546–550. doi: 10.1038/nrurol.2013.149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Lloyd GL, Ricke WA, McVary KT. Inflammation, voiding and benign prostatic hyperplasia progression. J Urol. 2019;201:868–870. doi: 10.1097/JU.0000000000000049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Macoska JA, Uchtmann KS, Leverson GE, McVary KT, Ricke WA. Prostate transition zone fibrosis is associated with clinical progression in the MTOPS study. J Urol. 2019;202:1240–1247. doi: 10.1097/JU.0000000000000385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Wynn TA, Ramalingam TR. Mechanisms of fibrosis: therapeutic translation for fibrotic disease. Nat Med. 2012;18:1028–1040. doi: 10.1038/nm.2807. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Vickman RE, Aaron-Brooks L, Zhang R, Lanman NA, Lapin B, Gil V, Greenberg M, Sasaki T, Cresswell GM, Broman MM, Paez JS, Petkewicz J, Talaty P, Helfand BT, Glaser AP, Wang CH, Franco OE, Ratliff TL, Nastiuk KL, Crawford SE, Hayward SW. TNF is a potential therapeutic target to suppress prostatic inflammation and hyperplasia in autoimmune disease. Nat Commun. 2022;13:2133. doi: 10.1038/s41467-022-29719-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Middleton LW, Shen Z, Varma S, Pollack AS, Gong X, Zhu S, Zhu C, Foley JW, Vennam S, Sweeney RT, Tu K, Biscocho J, Eminaga O, Nolley R, Tibshirani R, Brooks JD, West RB, Pollack JR. Genomic analysis of benign prostatic hyperplasia implicates cellular re-landscaping in disease pathogenesis. JCI Insight. 2019;5:e129749. doi: 10.1172/jci.insight.129749. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Nicholson TM, Ricke EA, Marker PC, Miano JM, Mayer RD, Timms BG, vom Saal FS, Wood RW, Ricke WA. Testosterone and 17beta-estradiol induce glandular prostatic growth, bladder outlet obstruction, and voiding dysfunction in male mice. Endocrinology. 2012;153:5556–5565. doi: 10.1210/en.2012-1522. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Begley L, Monteleon C, Shah RB, Macdonald JW, Macoska JA. CXCL12 overexpression and secretion by aging fibroblasts enhance human prostate epithelial proliferation in vitro. Aging Cell. 2005;4:291–298. doi: 10.1111/j.1474-9726.2005.00173.x. [DOI] [PubMed] [Google Scholar]
  • 12.Patalano S, Rodriguez-Nieves J, Colaneri C, Cotellessa J, Almanza D, Zhilin-Roth A, Riley T, Macoska J. CXCL12/CXCR4-mediated procollagen secretion is coupled to cullin-ring ubiquitin ligase activation. Sci Rep. 2018;8:3499. doi: 10.1038/s41598-018-21506-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Rodriguez-Nieves JA, Patalano SC, Almanza D, Gharaee-Kermani M, Macoska JA. CXCL12/CXCR4 axis activation mediates prostate myofibroblast phenoconversion through non-canonical EGFR/MEK/ERK signaling. PLoS One. 2016;11:e0159490. doi: 10.1371/journal.pone.0159490. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Macoska JA, Wang Z, Virta J, Zacharias N, Bjorling DE. Inhibition of the CXCL12/CXCR4 axis prevents periurethral collagen accumulation and lower urinary tract dysfunction in vivo. Prostate. 2019;79:757–767. doi: 10.1002/pros.23781. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.D’Arcy Q, Gharaee-Kermani M, Zhilin-Roth A, Macoska JA. The IL-4/IL-13 signaling axis promotes prostatic fibrosis. PLoS One. 2022;17:e0275064. doi: 10.1371/journal.pone.0275064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Joseph DB, Henry GH, Malewska A, Reese JC, Mauck RJ, Gahan JC, Hutchinson RC, Malladi VS, Roehrborn CG, Vezina CM, Strand DW. Single-cell analysis of mouse and human prostate reveals novel fibroblasts with specialized distribution and microenvironment interactions. J Pathol. 2021;255:141–154. doi: 10.1002/path.5751. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Joseph DB, Henry GH, Malewska A, Iqbal NS, Ruetten HM, Turco AE, Abler LL, Sandhu SK, Cadena MT, Malladi VS, Reese JC, Mauck RJ, Gahan JC, Hutchinson RC, Roehrborn CG, Baker LA, Vezina CM, Strand DW. Urethral luminal epithelia are castration-insensitive cells of the proximal prostate. Prostate. 2020;80:872–884. doi: 10.1002/pros.24020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Henry GH, Malewska A, Joseph DB, Malladi VS, Lee J, Torrealba J, Mauck RJ, Gahan JC, Raj GV, Roehrborn CG, Hon GC, MacConmara MP, Reese JC, Hutchinson RC, Vezina CM, Strand DW. A cellular anatomy of the normal adult human prostate and prostatic urethra. Cell Rep. 2018;25:3530–3542. e5. doi: 10.1016/j.celrep.2018.11.086. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Shoemaker R, Kim J. Urobiome: an outlook on the metagenome of urological diseases. Investig Clin Urol. 2021;62:611–622. doi: 10.4111/icu.20210312. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Kogan MI, Naboka YL, Ibishev KS, Gudima IA, Naber KG. Human urine is not sterile - shift of paradigm. Urol Int. 2015;94:445–452. doi: 10.1159/000369631. [DOI] [PubMed] [Google Scholar]
  • 21.Kogan MI, Naboka IuL, Ibishev KhS, Gudima IA. Unsterile urine in health human--new paradigm in medicine. Urologiia. 2014:48–52. [PubMed] [Google Scholar]
  • 22.Perez-Carrasco V, Soriano-Lerma A, Soriano M, Gutierrez-Fernandez J, Garcia-Salcedo JA. Urinary microbiome: yin and yang of the urinary tract. Front Cell Infect Microbiol. 2021;11:617002. doi: 10.3389/fcimb.2021.617002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Ozturk R, Murt A. Epidemiology of urological infections: a global burden. World J Urol. 2020;38:2669–2679. doi: 10.1007/s00345-019-03071-4. [DOI] [PubMed] [Google Scholar]
  • 24.Floyd KA, Moore JL, Eberly AR, Good JA, Shaffer CL, Zaver H, Almqvist F, Skaar EP, Caprioli RM, Hadjifrangiskou M. Adhesive fiber stratification in uropathogenic Escherichia coli biofilms unveils oxygen-mediated control of type 1 pili. PLoS Pathog. 2015;11:e1004697. doi: 10.1371/journal.ppat.1004697. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Starcic Erjavec M, Jesenicnik K, Elam LP, Kastrin A, Predojevic L, Sysoeva TA. Complete sequence of classic F-type plasmid pRK100 shows unique conservation over time and geographic location. Plasmid. 2022;119-120:102618. doi: 10.1016/j.plasmid.2022.102618. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Krewulak KD, Vogel HJ. Structural biology of bacterial iron uptake. Biochim Biophys Acta. 2008;1778:1781–1804. doi: 10.1016/j.bbamem.2007.07.026. [DOI] [PubMed] [Google Scholar]
  • 27.Bao GH, Ho CT, Barasch J. The ligands of neutrophil gelatinase-associated lipocalin. RSC Adv. 2015;5:104363–104374. doi: 10.1039/C5RA18736B. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Park MD, Silvin A, Ginhoux F, Merad M. Macrophages in health and disease. Cell. 2022;185:4259–4279. doi: 10.1016/j.cell.2022.10.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Mass E, Ballesteros I, Farlik M, Halbritter F, Gunther P, Crozet L, Jacome-Galarza CE, Handler K, Klughammer J, Kobayashi Y, Gomez-Perdiguero E, Schultze JL, Beyer M, Bock C, Geissmann F. Specification of tissue-resident macrophages during organogenesis. Science. 2016;353:aaf4238. doi: 10.1126/science.aaf4238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Wang AS, Steers NJ, Parab AR, Gachon F, Sweet MJ, Mysorekar IU. Timing is everything: impact of development, ageing and circadian rhythm on macrophage functions in urinary tract infections. Mucosal Immunol. 2022;15:1114–1126. doi: 10.1038/s41385-022-00558-z. [DOI] [PubMed] [Google Scholar]
  • 31.Jafari NV, Rohn JL. The urothelium: a multi-faceted barrier against a harsh environment. Mucosal Immunol. 2022;15:1127–1142. doi: 10.1038/s41385-022-00565-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Wiessner GB, Plumber SA, Xiang T, Mendelsohn CL. Development, regeneration and tumorigenesis of the urothelium. Development. 2022;149:dev198184. doi: 10.1242/dev.198184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Wang J, Batourina E, Schneider K, Souza S, Swayne T, Liu C, George CD, Tate T, Dan H, Wiessner G, Zhuravlev Y, Canman JC, Mysorekar IU, Mendelsohn CL. Polyploid superficial cells that maintain the urothelial barrier are produced via incomplete cytokinesis and endoreplication. Cell Rep. 2018;25:464–477. e4. doi: 10.1016/j.celrep.2018.09.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Liu C, Tate T, Batourina E, Truschel ST, Potter S, Adam M, Xiang T, Picard M, Reiley M, Schneider K, Tamargo M, Lu C, Chen X, He J, Kim H, Mendelsohn CL. Pparg promotes differentiation and regulates mitochondrial gene expression in bladder epithelial cells. Nat Commun. 2019;10:4589. doi: 10.1038/s41467-019-12332-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Ligon MM, Joshi CS, Fashemi BE, Salazar AM, Mysorekar IU. Effects of aging on urinary tract epithelial homeostasis and immunity. Dev Biol. 2023;493:29–39. doi: 10.1016/j.ydbio.2022.11.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Ligon MM, Wang C, DeJong EN, Schulz C, Bowdish DME, Mysorekar IU. Single cell and tissue-transcriptomic analysis of murine bladders reveals age- and TNFalpha-dependent but microbiota-independent tertiary lymphoid tissue formation. Mucosal Immunol. 2020;13:908–918. doi: 10.1038/s41385-020-0290-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Miyazato M, Yoshimura N, Chancellor MB. The other bladder syndrome: underactive bladder. Rev Urol. 2013;15:11–22. [PMC free article] [PubMed] [Google Scholar]
  • 38.Hughes FM Jr, Hirshman NA, Inouye BM, Jin H, Stanton EW, Yun CE, Davis LG, Routh JC, Purves JT. NLRP3 promotes diabetic bladder dysfunction and changes in symptom-specific bladder innervation. Diabetes. 2019;68:430–440. doi: 10.2337/db18-0845. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Hughes FM Jr, Odom MR, Cervantes A, Purves JT. Inflammation triggered by the NLRP3 inflammasome is a critical driver of diabetic bladder dysfunction. Front Physiol. 2022;13:920487. doi: 10.3389/fphys.2022.920487. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Hughes FM Jr, Allkanjari A, Odom MR, Jin H, Purves JT. Diabetic bladder dysfunction progresses from an overactive to an underactive phenotype in a type-1 diabetic mouse model (Akita female mouse) and is dependent on NLRP3. Life Sci. 2022;299:120528. doi: 10.1016/j.lfs.2022.120528. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Verbitsky M, Krithivasan P, Batourina E, Khan A, Graham SE, Marasà M, Kim H, Lim TY, Weng PL, Sánchez-Rodríguez E, Mitrotti A, Ahram DF, Zanoni F, Fasel DA, Westland R, Sampson MG, Zhang JY, Bodria M, Kil BH, Shril S, Gesualdo L, Torri F, Scolari F, Izzi C, van Wijk JAE, Saraga M, Santoro D, Conti G, Barton DE, Dobson MG, Puri P, Furth SL, Warady BA, Pisani I, Fiaccadori E, Allegri L, Degl’Innocenti ML, Piaggio G, Alam S, Gigante M, Zaza G, Esposito P, Lin F, Simões-E-Silva AC, Brodkiewicz A, Drozdz D, Zachwieja K, Miklaszewska M, Szczepanska M, Adamczyk P, Tkaczyk M, Tomczyk D, Sikora P, Mizerska-Wasiak M, Krzemien G, Szmigielska A, Zaniew M, Lozanovski VJ, Gucev Z, Ionita-Laza I, Stanaway IB, Crosslin DR, Wong CS, Hildebrandt F, Barasch J, Kenny EE, Loos RJF, Levy B, Ghiggeri GM, Hakonarson H, Latos-Bieleńska A, Materna-Kiryluk A, Darlow JM, Tasic V, Willer C, Kiryluk K, Sanna-Cherchi S, Mendelsohn CL, Gharavi AG. Copy number variant analysis and genome-wide association study identify loci with large effect for vesicoureteral reflux. J Am Soc Nephrol. 2021;32:805–820. doi: 10.1681/ASN.2020050681. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Verbitsky M, Westland R, Perez A, Kiryluk K, Liu Q, Krithivasan P, Mitrotti A, Fasel DA, Batourina E, Sampson MG, Bodria M, Werth M, Kao C, Martino J, Capone VP, Vivante A, Shril S, Kil BH, Marasà M, Zhang JY, Na YJ, Lim TY, Ahram D, Weng PL, Heinzen EL, Carrea A, Piaggio G, Gesualdo L, Manca V, Masnata G, Gigante M, Cusi D, Izzi C, Scolari F, van Wijk JAE, Saraga M, Santoro D, Conti G, Zamboli P, White H, Drozdz D, Zachwieja K, Miklaszewska M, Tkaczyk M, Tomczyk D, Krakowska A, Sikora P, Jarmoliński T, Borszewska-Kornacka MK, Pawluch R, Szczepanska M, Adamczyk P, Mizerska-Wasiak M, Krzemien G, Szmigielska A, Zaniew M, Dobson MG, Darlow JM, Puri P, Barton DE, Furth SL, Warady BA, Gucev Z, Lozanovski VJ, Tasic V, Pisani I, Allegri L, Rodas LM, Campistol JM, Jeanpierre C, Alam S, Casale P, Wong CS, Lin F, Miranda DM, Oliveira EA, Simões-E-Silva AC, Barasch JM, Levy B, Wu N, Hildebrandt F, Ghiggeri GM, Latos-Bielenska A, Materna-Kiryluk A, Zhang F, Hakonarson H, Papaioannou VE, Mendelsohn CL, Gharavi AG, Sanna-Cherchi S. The copy number variation landscape of congenital anomalies of the kidney and urinary tract. Nat Genet. 2019;51:117–127. doi: 10.1038/s41588-018-0281-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Cho SY, Bae J, Yoo C, Oh SJ. Establishment of a grading system for bladder trabeculation. Urology. 2013;81:503–507. doi: 10.1016/j.urology.2012.11.041. [DOI] [PubMed] [Google Scholar]
  • 44.Jung JH, Cho SY, Yoo C, Oh SJ. Establishment of the novel cystoscopic classification for bladder trabeculation of neurogenic bladder. Urology. 2014;84:515–519. doi: 10.1016/j.urology.2014.05.015. [DOI] [PubMed] [Google Scholar]
  • 45.Leslie SW, Tadi P, Tayyeb M. Neurogenic bladder and neurogenic lower urinary tract dysfunction. Treasure Island (FL): StatPearls; 2022. [PubMed] [Google Scholar]
  • 46.Heppner TJ, Tykocki NR, Hill-Eubanks D, Nelson MT. Transient contractions of urinary bladder smooth muscle are drivers of afferent nerve activity during filling. J Gen Physiol. 2016;147:323–335. doi: 10.1085/jgp.201511550. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Sant GR, Theoharides TC. The role of the mast cell in interstitial cystitis. Urol Clin North Am. 1994;21:41–53. [PubMed] [Google Scholar]
  • 48.Crawford LK, Caterina MJ. Functional anatomy of the sensory nervous system: updates from the neuroscience bench. Toxicol Pathol. 2020;48:174–189. doi: 10.1177/0192623319869011. [DOI] [PubMed] [Google Scholar]
  • 49.Wayman GA, Bose DD, Yang D, Lesiak A, Bruun D, Impey S, Ledoux V, Pessah IN, Lein PJ. PCB-95 modulates the calcium-dependent signaling pathway responsible for activity-dependent dendritic growth. Environ Health Perspect. 2012;120:1003–1009. doi: 10.1289/ehp.1104833. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Keil Stietz KP, Sethi S, Klocke CR, de Ruyter TE, Wilson MD, Pessah IN, Lein PJ. Sex and genotype modulate the dendritic effects of developmental exposure to a human-relevant polychlorinated biphenyls mixture in the juvenile mouse. Front Neurosci. 2021;15:766802. doi: 10.3389/fnins.2021.766802. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Keil Stietz KP, Kennedy CL, Sethi S, Valenzuela A, Nunez A, Wang K, Wang Z, Wang P, Spiegelhoff A, Puschner B, Bjorling DE, Lein PJ. In utero and lactational PCB exposure drives anatomic changes in the juvenile mouse bladder. Curr Res Toxicol. 2021;2:1–18. doi: 10.1016/j.crtox.2021.01.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Huang AJ, Brown JS, Thom DH, Fink HA, Yaffe K Study of Osteoporotic Fractures Research Group. Urinary incontinence in older community-dwelling women: the role of cognitive and physical function decline. Obstet Gynecol. 2007;109:909–916. doi: 10.1097/01.AOG.0000258277.01497.4b. [DOI] [PubMed] [Google Scholar]
  • 53.Suskind AM, Cawthon PM, Nakagawa S, Subak LL, Reinders I, Satterfield S, Cummings S, Cauley JA, Harris T, Huang AJ Health ABC Study. Urinary incontinence in older women: the role of body composition and muscle strength: from the health, aging, and body composition study. J Am Geriatr Soc. 2017;65:42–50. doi: 10.1111/jgs.14545. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Weaver JK, Milford K, Rickard M, Logan J, Erdman L, Viteri B, D’Souza N, Cucchiara A, Skreta M, Keefe D, Shah S, Selman A, Fischer K, Weiss DA, Long CJ, Lorenzo A, Fan Y, Tasian GE. Deep learning imaging features derived from kidney ultrasounds predict chronic kidney disease progression in children with posterior urethral valves. Pediatr Nephrol. 2022 doi: 10.1007/s00467-022-05677-0. [Epub ahead of print] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Yin S, Peng Q, Li H, Zhang Z, You X, Fischer K, Furth SL, Fan Y, Tasian GE. Multi-instance deep learning of ultrasound imaging data for pattern classification of congenital abnormalities of the kidney and urinary tract in children. Urology. 2020;142:183–189. doi: 10.1016/j.urology.2020.05.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Yin S, Peng Q, Li H, Zhang Z, You X, Fischer K, Furth SL, Tasian GE, Fan Y. Automatic kidney segmentation in ultrasound images using subsequent boundary distance regression and pixelwise classification networks. Med Image Anal. 2020;60:101602. doi: 10.1016/j.media.2019.101602. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Zheng Q, Furth SL, Tasian GE, Fan Y. Computer-aided diagnosis of congenital abnormalities of the kidney and urinary tract in children based on ultrasound imaging data by integrating texture image features and deep transfer learning image features. J Pediatr Urol. 2019;15:75.e1–75.e7. doi: 10.1016/j.jpurol.2018.10.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Babajide R, Lembrikova K, Ziemba J, Ding J, Li Y, Fermin AS, Fan Y, Tasian GE. Automated machine learning segmentation and measurement of urinary stones on CT scan. Urology. 2022;169:41–46. doi: 10.1016/j.urology.2022.07.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Ho DS, Scialabba D, Terry RS, Ma X, Chen J, Sankin GN, Xiang G, Qi R, Preminger GM, Lipkin ME, Zhong P. The role of cavitation in energy delivery and stone damage during laser lithotripsy. J Endourol. 2021;35:860–870. doi: 10.1089/end.2020.0349. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Winship B, Wollin D, Carlos E, Peters C, Li J, Terry R, Boydston K, Preminger GM, Lipkin ME. The rise and fall of high temperatures during ureteroscopic holmium laser lithotripsy. J Endourol. 2019;33:794–799. doi: 10.1089/end.2019.0084. [DOI] [PubMed] [Google Scholar]
  • 61.Terry RS, Whelan PS, Lipkin ME. New devices for kidney stone management. Curr Opin Urol. 2020;30:144–148. doi: 10.1097/MOU.0000000000000710. [DOI] [PubMed] [Google Scholar]
  • 62.Chen J, Li D, Yu W, Ma Z, Li C, Xiang G, Wu Y, Yao J, Zhong P. The effects of scanning speed and standoff distance of the fiber on dusting efficiency during short pulse holmium: YAG laser lithotripsy. J Clin Med. 2022;11:5048. doi: 10.3390/jcm11175048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Sierra A, Corrales M, Pinero A, Kolvatzis M, Somani B, Traxer O. Glossary of pre-settings given by laser companies: no consensus! World J Urol. 2022;40:2313–2321. doi: 10.1007/s00345-022-04090-4. [DOI] [PubMed] [Google Scholar]
  • 64.Okhunov Z, Jiang P, Afyouni AS, Ayad M, Arada R, Brevik A, Akopian G, Patel RM, Landman J, Clayman RV. Caveat emptor: the heat is “on”-an in vivo evaluation of the thulium fiber laser and temperature changes in the porcine kidney during dusting and fragmentation modes. J Endourol. 2021;35:1716–1722. doi: 10.1089/end.2021.0206. [DOI] [PubMed] [Google Scholar]
  • 65.Ziemba JB, Matlaga BR. Epidemiology and economics of nephrolithiasis. Investig Clin Urol. 2017;58:299–306. doi: 10.4111/icu.2017.58.5.299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Crivelli JJ, Mitchell T, Knight J, Wood KD, Assimos DG, Holmes RP, Fargue S. Contribution of dietary oxalate and oxalate precursors to urinary oxalate excretion. Nutrients. 2020;13:62. doi: 10.3390/nu13010062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Fargue S, Milliner DS, Knight J, Olson JB, Lowther WT, Holmes RP. Hydroxyproline metabolism and oxalate synthesis in primary hyperoxaluria. J Am Soc Nephrol. 2018;29:1615–1623. doi: 10.1681/ASN.2017040390. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Buchalski B, Wood KD, Challa A, Fargue S, Holmes RP, Lowther WT, Knight J. The effects of the inactivation of Hydroxyproline dehydrogenase on urinary oxalate and glycolate excretion in mouse models of primary hyperoxaluria. Biochim Biophys Acta Mol Basis Dis. 2020;1866:165633. doi: 10.1016/j.bbadis.2019.165633. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from American Journal of Clinical and Experimental Urology are provided here courtesy of e-Century Publishing Corporation

RESOURCES