Network


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

Hotspot


Dive into the research topics where Rehan Villani is active.

Publication


Featured researches published by Rehan Villani.


Blood | 2009

The Hedgehog receptor Patched1 regulates myeloid and lymphoid progenitors by distinct cell-extrinsic mechanisms.

Sarah Siggins; Nhu-Y Nguyen; Matthew P. McCormack; Sumitha Vasudevan; Rehan Villani; Stephen M. Jane; Brandon J. Wainwright; David J. Curtis

Hedgehog (Hh) ligands bind to the Patched1 (Ptch1) receptor, relieving repression of Smoothened, which leads to activation of the Hh signaling pathway. Using conditional Ptch1 knockout mice, the aim of this study was to determine the effects of activating the Hh signaling pathway in hematopoiesis. Surprisingly, hematopoietic-specific deletion of Ptch1 did not lead to activation of the Hh signaling pathway and, consequently, had no phenotypic effect. In contrast, deletion of Ptch1 in nonhematopoietic cells produced 2 distinct hematopoietic phenotypes. First, activation of Hh signaling in epithelial cells led to apoptosis of lymphoid progenitors associated with markedly elevated levels of circulating thymic stromal lymphopoietin. Second, activation of Hh signaling in the bone marrow cell niche led to increased numbers of lineage-negative c-kit(+) Sca-1(+) bone marrow cells and mobilization of myeloid progenitors associated with a marked loss of osteoblasts. Thus, deletion of Ptch1 leads to hematopoietic effects by distinct cell-extrinsic mechanisms rather than by direct activation of the Hh signaling pathway in hematopoietic cells. These findings have important implications for therapeutics designed to activate the Hh signaling pathway in hematopoietic cells including hematopoietic stem cells.


Cancer Prevention Research | 2010

Patched1 inhibits epidermal progenitor cell expansion and basal cell carcinoma formation by limiting Igfbp2 activity

Rehan Villani; Christelle Adolphe; James S. Palmer; Michael J. Waters; Brandon J. Wainwright

Basal cell carcinoma (BCC) of the skin is the most common form of cancer, with the majority being caused by mutations in the Patched1 (Ptch1) gene, leading to activation of the Hedgehog (Hh) signaling pathway. Hh signaling is implicated in many tumor types; thus, defining the mechanisms by which Ptch1 regulates tissue proliferation is of paramount importance. Here, we show that the key role of Ptch1 in the skin is to limit the size of the epidermal stem/progenitor compartment and allow hair follicle differentiation. Specifically, loss of Ptch1 leads to the promotion of progenitor cell fate by increasing basal cell proliferation and limiting the progression of basal cells into differentiated hair follicle cell types. Our data indicate that BCCs likely result from hair follicle progenitor cells that, due to Hh signal activation, cannot progress through normal hair follicle differentiation. These data confirm the role of Ptch1 as a negative regulator of epidermal progenitor turnover and also show for the first time that Ptch1 plays a role in the differentiation of the hair follicle lineage. In addition, we show that insulin-like growth factor binding protein 2 (Igfbp2) is upregulated in both murine and human BCCs and that blocking Igfbp2 activity reduces the Hh-mediated expansion of epidermal progenitor cells. We propose that Igfbp2 mediates epidermal progenitor cell expansion and therefore represents an epidermal progenitor cell–specific target of Hh signaling that promotes BCC development. Cancer Prev Res; 3(10); 1222–34. ©2010 AACR.


Journal of Investigative Dermatology | 2014

Patched 1 and Patched 2 Redundancy Has a Key Role in Regulating Epidermal Differentiation

Christelle Adolphe; Erica Nieuwenhuis; Rehan Villani; Zhu Juan Li; Pritinder Kaur; Chi-chung Hui; Brandon J. Wainwright

The Patched 1 (Ptch1) receptor has a pivotal role in inhibiting the activity of the Hedgehog (Hh) pathway and is therefore critical in preventing the onset of many human developmental disorders and tumor formation. However, the functional role of the mammalian Ptch2 paralogue remains elusive, particularly the extent to which it contributes to regulating the spatial and temporal activity of Hh signaling. Here we demonstrate in three independent mouse models of epidermal development that in vivo ablation of both Ptch receptors results in a more severe phenotype than loss of Ptch1 alone. Our studies indicate that concomitant loss of Ptch1 and Ptch2 activity inhibits epidermal lineage specification and differentiation. These results reveal that repression of Hh signaling through a dynamic Ptch regulatory network is a crucial event in lineage fate determination in the skin. In general, our findings implicate Ptch receptor redundancy as a key issue in elucidating the cellular origin of Hh-induced tumors.


Journal of Investigative Dermatology | 2014

Transgenic Flash Mice for In Vivo Quantitative Monitoring of Canonical Wnt Signaling to Track Hair Follicle Cycle Dynamics

Samantha Hodgson; Zoltán Neufeld; Rehan Villani; Edwige Roy; Kiarash Khosrotehrani

Hair follicles (HFs) upon development enter a lifelong cycle of growth, regression, and resting. These phases have been extensively studied at the cellular and molecular levels for individual HFs. However, HFs group into domains with coordinated cycling strongly influenced by their environment. These macroscopic hair domains have been difficult to study and can be influenced by physiological or pathological conditions, such as pregnancy or skin wounds. To robustly address this issue, we generated a mouse model for quantitative monitoring of β-catenin activity reflecting HF cycle dynamics macroscopically by using live bioluminescence imaging. These mice allowed live tracking of HF cycles and development, and highlighted hair regenerative patterns known to occur through macro-environmental cues, including initiation events, propagating anagen and border stability, and allowed refinement of a mechanistic mathematical model that integrates epidermal cell population dynamics into an excitable reaction-diffusion model. HF cycling could be studied in situations of pregnancy, wound healing, hair plucking, as well as in response to cyclosporine or Wnt3a stimulation. In conclusion, we developed a model for analysis of HF cycling at the macroscopic level that will allow refined analysis of hair cycle kinetics as well as its propagation dynamics.


Journal of Investigative Dermatology | 2017

Keratinocyte Sonic Hedgehog Up-regulation Drives the Development of Giant Congenital Nevi via Paracrine Endothelin-1 Secretion

Arash Chitsazan; Blake Ferguson; Rehan Villani; Herlina Y. Handoko; Pamela Mukhopadhyay; Brian Gabrielli; Wolter J. Mooi; H. Peter Soyer; Duncan Lambie; Kiarash Khosrotehrani; Grant Morahan; Graeme J. Walker

Giant congenital nevi are associated with clinical complications such as neurocutaneous melanosis and melanoma. Virtually nothing is known about why some individuals develop these lesions. We previously identified the sonic hedgehog (Shh) pathway regulator Cdon as a candidate nevus modifier gene. Here we validate this by studying Cdon knockout mice, and go on to establishing the mechanism by which Shh exacerbates nevogenesis. Cdon knockout mice develop blue nevi without the need for somatic melanocyte oncogenic mutation. In a mouse model carrying melanocyte NRASQ61K, we found that strain backgrounds that carry genetic variants that cause increased keratinocyte Shh pathway activity, as measured by Gli1 and Gli2 expression, develop giant congenital nevi. Shh components are also active adjacent to human congenital nevi. Mechanistically, this exacerbation of nevogenesis is driven via the release of the melanocyte mitogen endothelin-1 from keratinocytes. We then suppressed nevus development in mice using Shh and endothelin antagonists. Our work suggests an aspect of nevus development whereby keratinocyte cytokines such as endothelin-1 can exacerbate nevogenesis, and provides potential therapeutic approaches for giant congenital nevi. Furthermore, it highlights the notion that germline genetic variation, in addition to somatic melanocyte mutation, can strongly influence the histopathological features of melanocytic nevi.


Journal of Investigative Dermatology | 2014

Differential Effects of Ultraviolet Irradiation in Neonatal versus Adult Mice Are Not Explained by Defective Macrophage or Neutrophil Infiltration

Mathieu P. Rodero; Herlina Y. Handoko; Rehan Villani; Graeme J. Walker; Kiarash Khosrotehrani

Epidemiological studies suggest that ultraviolet B exposure (UVR) during childhood is the most important environmental risk factor for melanoma. In accordance, neonatal, but not adult, UVR exacerbates melanoma incidence in mouse models. The inability of neonates, as opposed to adults, to mount a proper neutrophil inflammatory response in the skin upon UVR exposure has been one of the driving hypotheses explaining this observation for the past decade. However, this aspect remains controversial. Here, we evaluated the UVR-induced inflammatory response in neonatal versus adult mice. In neonates, a significant neutrophil infiltration could be identified and quantified using three different antibodies by flow cytometry or immunohistochemistry. On day 1 after UVR, neutrophils were increased by 84-fold and on day 4 macrophages increased by 37-fold compared with nonexposed age-matched skin. When compared with adults, neonatal skin harbored a higher proportion of neutrophils in the myeloid compartment without significant differences in absolute counts. This response was reproduced with different kinetics in C57Bl/6 and FVB mice with a more rapid attenuation of neutrophil counts in the latter. Overall, our results suggest that the greatly increased sensitivity to melanomagenesis in neonates does not result from their incompetence in terms of myeloid inflammatory response to UVR.


Endocrine-related Cancer | 2013

Murine basal cell carcinoma leads to tumor-mediated alterations in endocrine Igf1 signaling.

Rehan Villani; Michael J Waters; Brandon J Wainwright

The intrinsic properties underlying cancer development are extensively studied while the effect of a cancer on the host is often overlooked. Activation of the Hedgehog (Hh) signaling pathway underlies a number of types of common human cancers, yet little is known concerning endocrine signaling in such tumors. Here, we investigated endocrine signaling in a murine model of basal cell carcinoma (BCC) of the skin, the most common cancer. BCCs were generated by the activation of Hh signaling resulting from the specific deletion of the Ptch1 gene in the developing epidermis. Subsequently, a severe growth deficiency was observed in the murine BCC model, and we identified a deficiency of circulating IGF1 (Igf1). We demonstrate that Hh pathway activation in murine BCC induces IGF binding proteins, thereby regulating Igf1 sequestration into the skin and skewing Igf endocrine signaling. Significantly, these results show that Hh-induced tumors can have endocrine effects on normal tissues that in turn can greatly impact the host. This study not only identifies that Igf is important in Hh-associated skin tumors but also exemplifies the need to consider endocrine signaling when interpreting complex in vivo tumor models.


Frontiers in Immunology | 2018

Temporal Regulation of Natural Killer T Cell Interferon Gamma Responses by β-Catenin-Dependent and -Independent Wnt Signaling

Jessica C. Kling; Margaret A. Jordan; Lauren A. Pitt; Jana Meiners; Thao Thanh-Tran; Le Son Tran; Tam T. K. Nguyen; Deepak Mittal; Rehan Villani; Raymond J. Steptoe; Kiarash Khosrotehrani; Stuart P. Berzins; Alan G. Baxter; Dale I. Godfrey; Antje Blumenthal

Natural killer T (NKT) cells are prominent innate-like lymphocytes in the liver with critical roles in immune responses during infection, cancer, and autoimmunity. Interferon gamma (IFN-γ) and IL-4 are key cytokines rapidly produced by NKT cells upon recognition of glycolipid antigens presented by antigen-presenting cells (APCs). It has previously been reported that the transcriptional coactivator β-catenin regulates NKT cell differentiation and functionally biases NKT cell responses toward IL-4, at the expense of IFN-γ production. β-Catenin is not only a central effector of Wnt signaling but also contributes to other signaling networks. It is currently unknown whether Wnt ligands regulate NKT cell functions. We thus investigated how Wnt ligands and β-catenin activity shape liver NKT cell functions in vivo in response to the glycolipid antigen, α-galactosylceramide (α-GalCer) using a mouse model. Pharmacologic targeting of β-catenin activity with ICG001, as well as myeloid-specific genetic ablation of Wntless (Wls), to specifically target Wnt protein release by APCs, enhanced early IFN-γ responses. By contrast, within several hours of α-GalCer challenge, myeloid-specific Wls deficiency, as well as pharmacologic targeting of Wnt release using the small molecule inhibitor IWP-2 impaired α-GalCer-induced IFN-γ responses, independent of β-catenin activity. These data suggest that myeloid cell-derived Wnt ligands drive early Wnt/β-catenin signaling that curbs IFN-γ responses, but that, subsequently, Wnt ligands sustain IFN-γ expression independent of β-catenin activity. Our analyses in ICG001-treated mice confirmed a role for β-catenin activity in driving early IL-4 responses by liver NKT cells. However, neither pharmacologic nor genetic perturbation of Wnt production affected the IL-4 response, suggesting that IL-4 production by NKT cells in response to α-GalCer is not driven by released Wnt ligands. Collectively, these data reveal complex temporal roles of Wnt ligands and β-catenin signaling in the regulation of liver NKT cell activation, and highlight Wnt-dependent and -independent contributions of β-catenin to NKT cell functions.


Development | 2017

Dominant-negative Sox18 function inhibits dermal papilla maturation and differentiation in all murine hair types

Rehan Villani; Samantha Hodgson; Julien M.D. Legrand; Jessica Greaney; Ho Yi Wong; Cathy Pichol-Thievend; Christelle Adolphe; Brandon Wainwight; Mathias Francois; Kiarash Khosrotehrani

SOX family proteins SOX2 and SOX18 have been reported as being essential in determining hair follicle type; however, the role they play during development remains unclear. Here, we demonstrate that Sox18 regulates the normal differentiation of the dermal papilla of all hair types. In guard (primary) hair dermal condensate (DC) cells, we identified transient Sox18 in addition to SOX2 expression at E14.5, which allowed fate tracing of primary DC cells until birth. Similarly, expression of Sox18 was detected in the DC cells of secondary hairs at E16.5 and in tertiary hair at E18.5. Dominant-negative Sox18 mutation (opposum) did not prevent DC formation in any hair type. However, it affected dermal papilla differentiation, restricting hair formation especially in secondary and tertiary hairs. This Sox18 mutation also prevented neonatal dermal cells or dermal papilla spheres from inducing hair in regeneration assays. Microarray expression studies identified WNT5A and TNC as potential downstream effectors of SOX18 that are important for epidermal WNT signalling. In conclusion, SOX18 acts as a mesenchymal molecular switch necessary for the formation and function of the dermal papilla in all hair types. Summary: The Sox18Op/+ mutation in mice, which mimics a hair loss condition in humans, leads to decreasing Wnt signal interactions with the hair follicle epidermis and abnormal hair follicle morphology.


British Journal of Dermatology | 2017

Oncogenes and morphogens: intricacies of targeted therapy in cutaneous basal cell carcinoma

Rehan Villani; Kiarash Khosrotehrani

Basal cell carcinomas (BCCs) are by far the most common malignancies in humans. This burden is mostly subsequent to field cancerization resulting in multiple primary tumours in the ultraviolet radiation-exposed field. This results in a chronic condition where life long surveillance and multiple excisions are needed. In a number of cases, BCCs are also capable of invasion and metastatic spread, justifying the development of targeted therapies such as hedgehog (Hh) pathway inhibitors. In this issue of the BJD, Dessinioti et al. undertook a comprehensive review of the Hh pathway and its role in hair follicle development and cycling, but also in BCC carcinogenesis. Historically, analysis of patients with Gorlin syndrome identified defective PTCH1 alleles as the causal gene leading to the activation of the Hh pathway. The importance of the Hh pathway in BCC is further confirmed through the study of patients with primary or secondary resistance to smoothened inhibitors where activation of the Hh pathway, as measured by GLI overexpression, is always observed. Besides its role in BCC formation, Hh signalling is also essential in hair follicle development and cycling. Even in oncogenic activation in the interfollicular epidermis, the Hh pathway induces a gene expression signature reminiscent of embryonic hair development processes. This explains to a large extent the shared morphology and keratin/ protein expression between BCCs and hair follicles. Although not discussed here, this similarity has been extended to the mesenchymal portion of hair follicles, the dermal papilla, resembling the stroma surrounding BCCs. Dessinioti et al. further detail the role of Hh signalling downstream of canonical Wnt signalling in hair follicle cycling and epidermal transit amplifying cell proliferation and differentiation in the hair matrix and the outer root sheath in the anagen phase. This is of importance to understand the alopeciaresembling telogen effluvium induced by Hh pathway inhibitors. Similarly, Hh ligand production by a perineural niche in the bulge of the hair follicle inducing the pathway directly and maintaining a subset of hair follicle stem cells could also be affected, thus modifying hair cycling and wound repair. Moreover, Hh signalling persists in the dermal papilla and maintains its identity, and it is well known that loss of dermal papilla signalling strongly affects hair follicle cycling. These observations might explain fibrous tract and loss of hair follicles over prolonged exposure to smoothened inhibitors, possibly explaining cases where regrowth of hair is not observed despite removal of Hh inhibitors. Overall, Dessinioti et al. remind us of the complexity of signalling pathways and their roles in physiological and pathological scenarios. It seems more and more an imperative for clinicians to fully embrace the molecular events that govern processes such as hair cycling and carcinogenesis for the adequate management of BCC. The intricacies of BCC-driving pathways and hair follicle cycling is a clear example of this and should be considered in developing further targeted therapies affecting Wnt or other key signalling pathways in this area.

Collaboration


Dive into the Rehan Villani's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Edwige Roy

University of Queensland

View shared research outputs
Top Co-Authors

Avatar

Graeme J. Walker

QIMR Berghofer Medical Research Institute

View shared research outputs
Top Co-Authors

Avatar

Herlina Y. Handoko

QIMR Berghofer Medical Research Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge