Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Nicolas F. Berbari is active.

Publication


Featured researches published by Nicolas F. Berbari.


Current Topics in Developmental Biology | 2008

Chapter 13 Ciliary Dysfunction in Developmental Abnormalities and Diseases

Neeraj Sharma; Nicolas F. Berbari; Bradley K. Yoder

Cilia are small microtubule-based cellular appendages that are broadly classified as being either motile or immotile (primary cilia). Since their initial discovery several centuries ago, motile cilia have been of general interest to basic scientists and others who study the dynamics and physiological relevance of their motility. More recent discoveries have found that motile and immotile cilia, the later of which are present on nearly all cells in the mammalian body, also have major roles during development and in postnatal life. Dysfunction of the cilium is the basis for multiple human genetic disorders that have collectively been called the ciliopathies. The phenotypes associated with cilia dysfunction in mammals are diverse and include randomization of the left-right body axis, abnormalities in neural tube closure and patterning, skeletal defects such as polydactyly, cystic kidney, liver, and pancreatic diseases, blindness and anosmia, behavioral and cognitive defects, and obesity. The connection between disease and developmental defects due to the loss of ciliary function has brought the efforts of the biomedical research establishment to bear on this underappreciated and long overlooked organelle. Several groups have applied en silico, genetic, and biochemical approaches to identify the components of the cilia proteome. The resulting datasets have contributed to a remarkable increase in the rate at which human ciliopathy disease loci are being identified. This intense basic and clinical research interest has revealed that the cilium is a very complex sensory machine involved in transducing extracellular stimuli involved in many different signaling pathways into cellular responses. Although major advances have been made in understanding the importance of the cilium, it remains enigmatic how the cilium functions to coordinate signaling pathways and how loss of this organelle results in the severe defects observed in human ciliopathies.


Molecular Biology of the Cell | 2011

Soluble levels of cytosolic tubulin regulate ciliary length control

Neeraj Sharma; Zachary A. Kosan; Jannese E. Stallworth; Nicolas F. Berbari; Bradley K. Yoder

We show that manipulation of either the microtubule or the actin cytoskeleton has unexpected influences on cilia length control.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Leptin resistance is a secondary consequence of the obesity in ciliopathy mutant mice

Nicolas F. Berbari; Raymond C. Pasek; Erik B. Malarkey; S. M. Zaki Yazdi; Andrew D. McNair; Wesley R. Lewis; Tim R. Nagy; Robert A. Kesterson; Bradley K. Yoder

Although primary cilia are well established as important sensory and signaling structures, their function in most tissues remains unknown. Obesity is a feature associated with some syndromes of cilia dysfunction, such as Bardet-Biedl syndrome (BBS) and Alström syndrome, as well as in several cilia mutant mouse models. Recent data indicate that obesity in BBS mutant mice is due to defects in leptin receptor trafficking and leptin resistance. Furthermore, induction of cilia loss in leptin-responsive proopiomelanocortin neurons results in obesity, implicating cilia on hypothalamic neurons in regulating feeding behavior. Here, we directly test the importance of the cilium as a mediator of the leptin response. In contrast to the current dogma, a longitudinal study of conditional Ift88 cilia mutant mice under different states of adiposity indicates that leptin resistance is present only when mutants are obese. Our studies show that caloric restriction leads to an altered anticipatory feeding behavior that temporarily abrogates the anorectic actions of leptin despite normalized circulating leptin levels. Interestingly, preobese Bbs4 mutant mice responded to the anorectic effects of leptin and did not display other phenotypes associated with defective leptin signaling. Furthermore, thermoregulation and activity measurements in cilia mutant mice are inconsistent with phenotypes previously observed in leptin deficient ob/ob mice. Collectively, these data indicate that cilia are not directly involved in leptin responses and that a defect in the leptin signaling axis is not the initiating event leading to hyperphagia and obesity associated with cilia dysfunction.


Developmental Biology | 2011

Mutations in Traf3ip1 reveal defects in ciliogenesis, embryonic development, and altered cell size regulation

Nicolas F. Berbari; Nicholas W. Kin; Neeraj Sharma; Edward J. Michaud; Robert A. Kesterson; Bradley K. Yoder

Tumor necrosis factor alpha receptor 3 interacting protein 1 (Traf3ip1), also known as MIPT3, was initially characterized through its interactions with tubulin, actin, TNFR-associated factor-3 (Traf3), IL-13R1, and DISC1. It functions as an inhibitor of IL-13-mediated phosphorylation of Stat6 and in sequestration of Traf3 and DISC1 to the cytoskeleton. Studies of the Traf3ip1 homologs in C. elegans (DYF-11), Zebrafish (elipsa), and Chlamydomonas (IFT54) revealed that the protein localizes to the cilium and is required for ciliogenesis. Similar localization data has now been reported for mammalian Traf3ip1. This raises the possibility that Traf3ip1 has an evolutionarily conserved role in mammalian ciliogenesis in addition to its previously indicated functions. To evaluate this possibility, a Traf3ip1 mutant mouse line was generated. Traf3ip1 mutant cells are unable to form cilia. Homozygous Traf3ip1 mutant mice are not viable and have both neural developmental defects and polydactyly, phenotypes typical of mouse mutants with ciliary assembly defects. Furthermore, in Traf3ip1 mutants the hedgehog pathway is disrupted, as evidenced by abnormal dorsal-ventral neural tube patterning and diminished expression of a hedgehog reporter. Analysis of the canonical Wnt pathway indicates that it was largely unaffected; however, specific domains in the pharyngeal arches have elevated levels of reporter activity. Interestingly, Traf3ip1 mutant embryos and cells failed to show alterations in IL-13 signaling, one of the pathways associated with its initial discovery. Novel phenotypes observed in Traf3ip1 mutant cells include elevated cytosolic levels of acetylated microtubules and a marked increase in cell size in culture. The enlarged Traf3ip1 mutant cell size was associated with elevated basal mTor pathway activity. Taken together, these data demonstrate that Traf3ip1 function is highly conserved in ciliogenesis and is important for proper regulation of a number of essential developmental and cellular pathways. The Traf3ip1 mutant mouse and cell lines will provide valuable resources to assess cilia function in mammalian development and also serve as a tool to explore the potential connections between cilia and cytoskeletal dynamics, mTor regulation, and cell volume control.


Cilia | 2013

An inducible CiliaGFP mouse model for in vivo visualization and analysis of cilia in live tissue

Amber K. O’Connor; Erik B. Malarkey; Nicolas F. Berbari; Mandy J. Croyle; Courtney J. Haycraft; P. Darwin Bell; Peter Hohenstein; Robert A. Kesterson; Bradley K. Yoder

BackgroundCilia are found on nearly every cell type in the mammalian body, and have been historically classified as either motile or immotile. Motile cilia are important for fluid and cellular movement; however, the roles of non-motile or primary cilia in most tissues remain unknown. Several genetic syndromes, called the ciliopathies, are associated with defects in cilia structure or function and have a wide range of clinical presentations. Much of what we know about the formation and maintenance of cilia comes from model systems like C. elegans and Chalmydomonas. Studies of mammalian cilia in live tissues have been hampered by difficulty visualizing them.ResultsTo facilitate analyses of mammalian cilia function we generated an inducible CiliaGFP mouse by targeting mouse cDNA encoding a cilia-localized protein somatostatin receptor 3 fused to GFP (Sstr3::GFP) into the ROSA26 locus. In this system, Sstr3::GFP is expressed from the ubiquitous ROSA26 promoter after Cre mediated deletion of an upstream Neo cassette flanked by lox P sites. Fluorescent cilia labeling was observed in a variety of live tissues and after fixation. Both cell-type specific and temporally regulated cilia labeling were obtained using multiple Cre lines. The analysis of renal cilia in anesthetized live mice demonstrates that cilia commonly lay nearly parallel to the apical surface of the tubule. In contrast, in more deeply anesthetized mice the cilia display a synchronized, repetitive oscillation that ceases upon death, suggesting a relationship to heart beat, blood pressure or glomerular filtration.ConclusionsThe ability to visualize cilia in live samples within the CiliaGFP mouse will greatly aid studies of ciliary function. This mouse will be useful for in vivo genetic and pharmacological screens to assess pathways regulating cilia motility, signaling, assembly, trafficking, resorption and length control and to study cilia regulated physiology in relation to ciliopathy phenotypes.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Primary cilia enhance kisspeptin receptor signaling on gonadotropin-releasing hormone neurons

Andrew Ian Koemeter-Cox; Thomas W. Sherwood; Jill A. Green; Robert A. Steiner; Nicolas F. Berbari; Bradley K. Yoder; Alexander S. Kauffman; Paula C. Monsma; Anthony Brown; Candice C. Askwith; Kirk Mykytyn

Significance Mammalian central neurons typically possess solitary appendages called primary cilia. These rod-shaped protrusions are enriched for signaling proteins, suggesting they respond to extracellular neuromodulators. Yet, neuronal ciliary signaling has never been observed and the roles of cilia in neuronal function are unclear. This study identifies Kiss1r, which plays a key role in reproductive function, as a novel ciliary receptor on gonadotropin-releasing hormone (GnRH) neurons. GnRH neurons possess multiple Kiss1r-positive cilia and the proportion of multiple cilia increases in parallel with pubertal maturation. Ablation of Kiss1r-positive cilia on GnRH neurons does not affect neuron migration or sexual maturation. However, kisspeptin-mediated increases in GnRH neuron firing rate are reduced in the absence of cilia. Thus, cilia enhance Kiss1r signaling on GnRH neurons. Most central neurons in the mammalian brain possess an appendage called a primary cilium that projects from the soma into the extracellular space. The importance of these organelles is highlighted by the fact that primary cilia dysfunction is associated with numerous neuropathologies, including hyperphagia-induced obesity, hypogonadism, and learning and memory deficits. Neuronal cilia are enriched for signaling molecules, including certain G protein-coupled receptors (GPCRs), suggesting that neuronal cilia sense and respond to neuromodulators in the extracellular space. However, the impact of cilia on signaling to central neurons has never been demonstrated. Here, we show that the kisspeptin receptor (Kiss1r), a GPCR that is activated by kisspeptin to regulate the onset of puberty and adult reproductive function, is enriched in cilia projecting from mouse gonadotropin-releasing hormone (GnRH) neurons. Interestingly, GnRH neurons in adult animals are multiciliated and the percentage of GnRH neurons possessing multiple Kiss1r-positive cilia increases during postnatal development in a progression that correlates with sexual maturation. Remarkably, disruption of cilia selectively on GnRH neurons leads to a significant reduction in kisspeptin-mediated GnRH neuronal activity. To our knowledge, this result is the first demonstration of cilia disruption affecting central neuronal activity and highlights the importance of cilia for proper GPCR signaling.


Journal of The American Society of Nephrology | 2013

Proximal Tubule Proliferation Is Insufficient to Induce Rapid Cyst Formation after Cilia Disruption

Neeraj Sharma; Erik B. Malarkey; Nicolas F. Berbari; Amber K. O’Connor; Gregory B. Vanden Heuvel; Michal Mrug; Bradley K. Yoder

Disrupting the function of cilia in mouse kidneys results in rapid or slow progression of cystic disease depending on whether the animals are juveniles or adults, respectively. Renal injury can also markedly accelerate the renal cyst formation that occurs after disruption of cilia in adult mice. Rates of cell proliferation are markedly higher in juvenile than adult kidneys and increase after renal injury, suggesting that cell proliferation may enhance the development of cysts. Here, we induced cilia loss in the kidneys of adult mice in the presence or absence of a Cux-1 transgene, which maintains cell proliferation. By using this model, we were able to avoid additional factors such as inflammation and dedifferentiation, which associate with renal injury and may also influence the rate of cystogenesis. After induction of cilia loss, cystic disease was not more pronounced in adult mice with the Cux-1 transgene compared with those without the transgene. In conclusion, these data suggest that proliferation is unlikely to be the sole mechanism underlying the rapid cystogenesis observed after injury in mice that lose cilia function in adulthood.


Cytoskeleton | 2013

Microtubule modifications and stability are altered by cilia perturbation and in cystic kidney disease

Nicolas F. Berbari; Neeraj Sharma; Erik B. Malarkey; Jay N. Pieczynski; Ravindra Boddu; Jacek Gaertig; Lisa M. Guay-Woodford; Bradley K. Yoder

Disruption of the primary cilium is associated with a growing number of human diseases collectively termed ciliopathies. Ciliopathies present with a broad range of clinical features consistent with the near ubiquitous nature of the organelle and its role in diverse signaling pathways throughout development and adult homeostasis. The clinical features associated with cilia dysfunction can include such phenotypes as polycystic kidneys, skeletal abnormalities, blindness, anosmia, and obesity. Although the clinical relevance of the primary cilium is evident, the effects that cilia dysfunction has on the cell and how this contributes to disease remains poorly understood. Here, we show that loss of ciliogenesis genes such as Ift88 and Kif3a lead to increases in post‐translational modifications on cytosolic microtubules. This effect was observed in cilia mutant kidney cells grown in vitro and in vivo in cystic kidneys. The hyper‐acetylation of microtubules resulting from cilia loss is associated with both altered microtubule stability and increased α‐tubulin acetyl‐transferase activity. Intriguingly, the effect on microtubules was also evident in renal samples from patients with autosomal recessive polycystic kidneys. These findings indicate that altered microtubule post‐translational modifications may influence some of the phenotypes observed in ciliopathies.


Cold Spring Harbor Perspectives in Biology | 2017

Cilia and Obesity

Christian Vaisse; Jeremy F. Reiter; Nicolas F. Berbari

The ciliopathies Bardet-Biedl syndrome and Alström syndrome cause obesity. How ciliary dysfunction leads to obesity has remained mysterious, partly because of a lack of understanding of the physiological roles of primary cilia in the organs and pathways involved in the regulation of metabolism and energy homeostasis. Historically, the study of rare monogenetic disorders that present with obesity has informed our molecular understanding of the mechanisms involved in nonsyndromic forms of obesity. Here, we present a framework, based on genetic studies in mice and humans, of the molecular and cellular pathways underlying long-term regulation of energy homeostasis. We focus on recent progress linking these pathways to the function of the primary cilia with a particular emphasis on the roles of neuronal primary cilia in the regulation of satiety.


PLOS ONE | 2014

Hippocampal and Cortical Primary Cilia Are Required for Aversive Memory in Mice

Nicolas F. Berbari; Erik B. Malarkey; S. M. Zaki Yazdi; Andrew D. McNair; Jordyn M. Kippe; Mandy J. Croyle; Timothy W. Kraft; Bradley K. Yoder

It has been known for decades that neurons throughout the brain possess solitary, immotile, microtubule based appendages called primary cilia. Only recently have studies tried to address the functions of these cilia and our current understanding remains poor. To determine if neuronal cilia have a role in behavior we specifically disrupted ciliogenesis in the cortex and hippocampus of mice through conditional deletion of the Intraflagellar Transport 88 (Ift88) gene. The effects on learning and memory were analyzed using both Morris Water Maze and fear conditioning paradigms. In comparison to wild type controls, cilia mutants displayed deficits in aversive learning and memory and novel object recognition. Furthermore, hippocampal neurons from mutants displayed an altered paired-pulse response, suggesting that loss of IFT88 can alter synaptic properties. A variety of other behavioral tests showed no significant differences between conditional cilia mutants and controls. This type of conditional allele approach could be used to distinguish which behavioral features of ciliopathies arise due to defects in neural development and which result from altered cell physiology. Ultimately, this could lead to an improved understanding of the basis for the cognitive deficits associated with human cilia disorders such as Bardet-Biedl syndrome, and possibly more common ailments including depression and schizophrenia.

Collaboration


Dive into the Nicolas F. Berbari's collaboration.

Top Co-Authors

Avatar

Bradley K. Yoder

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Erik B. Malarkey

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Robert A. Kesterson

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Neeraj Sharma

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Raymond C. Pasek

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Mandy J. Croyle

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Wesley R. Lewis

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Abraham L. Kierszenbaum

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Amber K. O’Connor

Children's National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Andrew D. McNair

University of Alabama at Birmingham

View shared research outputs
Researchain Logo
Decentralizing Knowledge