Network


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

Hotspot


Dive into the research topics where Richard Francis is active.

Publication


Featured researches published by Richard Francis.


Nature Genetics | 2013

DYX1C1 is required for axonemal dynein assembly and ciliary motility

Aarti Tarkar; Niki T. Loges; Christopher E. Slagle; Richard Francis; Gerard W. Dougherty; Joel V. Tamayo; Brett A. Shook; Marie E. Cantino; D. A. Schwartz; Charlotte Jahnke; Heike Olbrich; Claudius Werner; Johanna Raidt; Petra Pennekamp; Marouan Abouhamed; Rim Hjeij; Gabriele Köhler; Matthias Griese; You Li; Kristi Lemke; Nikolas Klena; Xiaoqin Liu; George C. Gabriel; Kimimasa Tobita; Martine Jaspers; Lucy Morgan; Adam J. Shapiro; Stef J.F. Letteboer; Dorus A. Mans; Johnny L. Carson

DYX1C1 has been associated with dyslexia and neuronal migration in the developing neocortex. Unexpectedly, we found that deleting exons 2–4 of Dyx1c1 in mice caused a phenotype resembling primary ciliary dyskinesia (PCD), a disorder characterized by chronic airway disease, laterality defects and male infertility. This phenotype was confirmed independently in mice with a Dyx1c1 c.T2A start-codon mutation recovered from an N-ethyl-N-nitrosourea (ENU) mutagenesis screen. Morpholinos targeting dyx1c1 in zebrafish also caused laterality and ciliary motility defects. In humans, we identified recessive loss-of-function DYX1C1 mutations in 12 individuals with PCD. Ultrastructural and immunofluorescence analyses of DYX1C1-mutant motile cilia in mice and humans showed disruptions of outer and inner dynein arms (ODAs and IDAs, respectively). DYX1C1 localizes to the cytoplasm of respiratory epithelial cells, its interactome is enriched for molecular chaperones, and it interacts with the cytoplasmic ODA and IDA assembly factor DNAAF2 (KTU). Thus, we propose that DYX1C1 is a newly identified dynein axonemal assembly factor (DNAAF4).


Circulation | 2012

High Prevalence of Respiratory Ciliary Dysfunction in Congenital Heart Disease Patients With Heterotaxy

Nader Nakhleh; Richard Francis; Rachel Giese; Xin Tian; You Li; Maimoona A. Zariwala; Hisato Yagi; Omar Khalifa; Safina Kureshi; Bishwanath Chatterjee; Steven L. Sabol; Matthew W. Swisher; Patricia S. Connelly; Matthew P. Daniels; Ashok Srinivasan; Karen Kuehl; Nadav Kravitz; Kimberlie A. Burns; Iman Sami; Heymut Omran; M. Michael Barmada; Kenneth N. Olivier; Kunal K. Chawla; Margaret W. Leigh; Richard A. Jonas; Linda Leatherbury; Cecilia W. Lo

Background— Patients with congenital heart disease (CHD) and heterotaxy show high postsurgical morbidity/mortality, with some developing respiratory complications. Although this finding is often attributed to the CHD, airway clearance and left-right patterning both require motile cilia function. Thus, airway ciliary dysfunction (CD) similar to that of primary ciliary dyskinesia (PCD) may contribute to increased respiratory complications in heterotaxy patients. Methods and Results— We assessed 43 CHD patients with heterotaxy for airway CD. Videomicrocopy was used to examine ciliary motion in nasal tissue, and nasal nitric oxide (nNO) was measured; nNO level is typically low with PCD. Eighteen patients exhibited CD characterized by abnormal ciliary motion and nNO levels below or near the PCD cutoff values. Patients with CD aged >6 years show increased respiratory symptoms similar to those seen in PCD. Sequencing of all 14 known PCD genes in 13 heterotaxy patients with CD, 12 without CD, 10 PCD disease controls, and 13 healthy controls yielded 0.769, 0.417, 1.0, and 0.077 novel variants per patient, respectively. One heterotaxy patient with CD had the PCD causing DNAI1 founder mutation. Another with hyperkinetic ciliary beat had 2 mutations in DNAH11, the only PCD gene known to cause hyperkinetic beat. Among PCD patients, 2 had known PCD causing CCDC39 and CCDC40 mutations. Conclusions— Our studies show that CHD patients with heterotaxy have substantial risk for CD and increased respiratory disease. Heterotaxy patients with CD were enriched for mutations in PCD genes. Future studies are needed to assess the potential benefit of prescreening and prophylactically treating heterotaxy patients for CD.


Developmental Cell | 2014

IFT27 Links the BBSome to IFT for Maintenance of the Ciliary Signaling Compartment

Thibaut Eguether; Jovenal T. San Agustin; Brian T. Keady; Julie A. Jonassen; Yinwen Liang; Richard Francis; Kimimasa Tobita; Colin A. Johnson; Zakia I.A. Abdelhamed; Cecilia W. Lo; Gregory J. Pazour

Vertebrate hedgehog signaling is coordinated by the differential localization of the receptors patched-1 and Smoothened in the primary cilium. Cilia assembly is mediated by intraflagellar transport (IFT), and cilia defects disrupt hedgehog signaling, causing many structural birth defects. We generated Ift25 and Ift27 knockout mice and show that they have structural birth defects indicative of hedgehog signaling dysfunction. Surprisingly, ciliary assembly is not affected, but abnormal hedgehog signaling is observed in conjunction with ciliary accumulation of patched-1 and Smoothened. Similarly, Smoothened accumulates in cilia on cells mutated for BBSome components or the BBS binding protein/regulator Lztfl1. Interestingly, the BBSome and Lztfl1 accumulate to high levels in Ift27 mutant cilia. Because Lztfl1 mutant cells accumulate BBSome but not IFT27, it is likely that Lztfl1 functions downstream of IFT27 to couple the BBSome to the IFT particle for coordinated removal of patched-1 and Smoothened from cilia during hedgehog signaling.


American Journal of Human Genetics | 2013

ARMC4 mutations cause primary ciliary dyskinesia with randomization of left/right body asymmetry.

Rim Hjeij; Anna Lindstrand; Richard Francis; Maimoona A. Zariwala; Xiaoqin Liu; You Li; Rama Rao Damerla; Gerard W. Dougherty; Marouan Abouhamed; Heike Olbrich; Niki T. Loges; Petra Pennekamp; Erica E. Davis; Claudia M.B. Carvalho; Davut Pehlivan; Claudius Werner; Johanna Raidt; Gabriele Köhler; Karsten Häffner; Miguel Reyes-Múgica; James R. Lupski; Margaret W. Leigh; Margaret Rosenfeld; Lucy Morgan; Cecilia W. Lo; Nicholas Katsanis; Heymut Omran

The motive forces for ciliary movement are generated by large multiprotein complexes referred to as outer dynein arms (ODAs), which are preassembled in the cytoplasm prior to transport to the ciliary axonemal compartment. In humans, defects in structural components, docking complexes, or cytoplasmic assembly factors can cause primary ciliary dyskinesia (PCD), a disorder characterized by chronic airway disease and defects in laterality. By using combined high resolution copy-number variant and mutation analysis, we identified ARMC4 mutations in twelve PCD individuals whose cells showed reduced numbers of ODAs and severely impaired ciliary beating. Transient suppression in zebrafish and analysis of an ENU mouse mutant confirmed in both model organisms that ARMC4 is critical for left-right patterning. We demonstrate that ARMC4 is an axonemal protein that is necessary for proper targeting and anchoring of ODAs.


PLOS ONE | 2011

Connexin43 Modulates Cell Polarity and Directional Cell Migration by Regulating Microtubule Dynamics

Richard Francis; Xin Xu; Hyunsoo Park; Chin-Jen Wei; Stephen Chang; Bishwanath Chatterjee; Cecilia W. Lo

Knockout mice deficient in the gap junction gene connexin43 exhibit developmental anomalies associated with abnormal neural crest, primordial germ cell, and proepicardial cell migration. These migration defects are due to a loss of directional cell movement, and are associated with abnormal actin stress fiber organization and a loss of polarized cell morphology. To elucidate the mechanism by which Cx43 regulates cell polarity, we used a wound closure assays with mouse embryonic fibroblasts (MEFs) to examine polarized cell morphology and directional cell movement. Studies using embryonic fibroblasts from Cx43 knockout (Cx43KO) mice showed Cx43 deficiency caused cell polarity defects as characterized by a failure of the Golgi apparatus and the microtubule organizing center to reorient with the direction of wound closure. Actin stress fibers at the wound edge also failed to appropriately align, and stabilized microtubule (Glu-tubulin) levels were markedly reduced. Forced expression of Cx43 with deletion of its tubulin-binding domain (Cx43dT) in both wildtype MEFs and neural crest cell explants recapitulated the cell migration defects seen in Cx43KO cells. However, forced expression of Cx43 with point mutation causing gap junction channel closure had no effect on cell motility. TIRF imaging revealed increased microtubule instability in Cx43KO cells, and microtubule targeting of membrane localized Cx43 was reduced with expression of Cx43dT construct in wildtype cells. Together, these findings suggest the essential role of Cx43 gap junctions in development is mediated by regulation of the tubulin cytoskeleton and cell polarity by Cx43 via a nonchannel function.


PLOS Biology | 2013

Wdpcp, a PCP Protein Required for Ciliogenesis, Regulates Directional Cell Migration and Cell Polarity by Direct Modulation of the Actin Cytoskeleton

Cheng Cui; Bishwanath Chatterjee; Thomas P. Lozito; Zhen Zhang; Richard Francis; Hisato Yagi; Lisa M. Swanhart; Subramaniam Sanker; Deanne Francis; Qing Yu; Jovenal T. San Agustin; Chandrakala Puligilla; Tania Chatterjee; Terry Tansey; Xiaoqin Liu; Matthew W. Kelley; Elias T. Spiliotis; Adam V. Kwiatkowski; Rocky S. Tuan; Gregory J. Pazour; Neil A. Hukriede; Cecilia W. Lo

Wdpcp, a protein required for both planar cell polarity and ciliogenesis, regulates cell polarity and alignment via direct modulation of the actin cytoskeleton.


Nature Genetics | 2015

Discovery of four recessive developmental disorders using probabilistic genotype and phenotype matching among 4,125 families.

Nadia A. Akawi; Jeremy McRae; Morad Ansari; Meena Balasubramanian; Moira Blyth; Angela F. Brady; Stephen Clayton; Trevor Cole; Charu Deshpande; Tomas Fitzgerald; Nicola Foulds; Richard Francis; George C. Gabriel; Sebastian S. Gerety; Judith A. Goodship; Emma Hobson; Wendy D Jones; Shelagh Joss; Daniel A. King; Nikolai T. Klena; Ajith Kumar; Melissa Lees; Chris Lelliott; Jenny Lord; Dominic McMullan; Mary O'Regan; Deborah Osio; Virginia Piombo; Elena Prigmore; Diana Rajan

Discovery of most autosomal recessive disease-associated genes has involved analysis of large, often consanguineous multiplex families or small cohorts of unrelated individuals with a well-defined clinical condition. Discovery of new dominant causes of rare, genetically heterogeneous developmental disorders has been revolutionized by exome analysis of large cohorts of phenotypically diverse parent-offspring trios. Here we analyzed 4,125 families with diverse, rare and genetically heterogeneous developmental disorders and identified four new autosomal recessive disorders. These four disorders were identified by integrating Mendelian filtering (selecting probands with rare, biallelic and putatively damaging variants in the same gene) with statistical assessments of (i) the likelihood of sampling the observed genotypes from the general population and (ii) the phenotypic similarity of patients with recessive variants in the same candidate gene. This new paradigm promises to catalyze the discovery of novel recessive disorders, especially those with less consistent or nonspecific clinical presentations and those caused predominantly by compound heterozygous genotypes.


American Journal of Physiology-heart and Circulatory Physiology | 2012

Congenital heart disease and the specification of left-right asymmetry

Richard Francis; Adam Christopher; William A. Devine; Lawrence E. Ostrowski; Cecilia W. Lo

Complex congenital heart disease (CHD) is often seen in conjunction with heterotaxy, the randomization of left-right visceral organ situs. However, the link between cardiovascular morphogenesis and left-right patterning is not well understood. To elucidate the role of left-right patterning in cardiovascular development, we examined situs anomalies and CHD in mice with a loss of function allele of Dnaic1, a dynein protein required for motile cilia function and left-right patterning. Dnaic1 mutants were found to have nodal cilia required for left-right patterning, but they were immotile. Half the mutants had concordant organ situs comprising situs solitus or mirror symmetric situs inversus. The remaining half had randomized organ situs or heterotaxy. Looping of the heart tube, the first anatomical lateralization, showed abnormal L-loop bias rather than the expected D-loop orientation in heterotaxy and nonheterotaxy mutants. Situs solitus/inversus mutants were viable with mild or no defects consisting of azygos continuation and/or ventricular septal defects, whereas all heterotaxy mutants had complex CHD. In heterotaxy mutants, but not situs solitus/inversus mutants, the morphological left ventricle was thin and often associated with a hypoplastic transverse aortic arch. Thus, in conclusion, Dnaic1 mutants can achieve situs solitus or inversus even with immotile nodal cilia. However, the finding of abnormal L-loop bias in heterotaxy and nonheterotaxy mutants would suggest motile cilia are required for normal heart looping. Based on these findings, we propose motile nodal cilia patterns heart looping but heart and visceral organ lateralization is driven by signaling not requiring nodal cilia motility.


The Journal of Thoracic and Cardiovascular Surgery | 2014

Increased postoperative respiratory complications in heterotaxy congenital heart disease patients with respiratory ciliary dysfunction

Brandon W. Harden; Xin Tian; Rachel Giese; Nader Nakhleh; Safina Kureshi; Richard Francis; Sridhar Hanumanthaiah; You Li; Matthew W. Swisher; Karen Kuehl; Iman Sami; Kenneth N. Olivier; Richard A. Jonas; Cecilia W. Lo; Linda Leatherbury

OBJECTIVE(S)nCongenital heart disease (CHD) and heterotaxy patients have increased postoperative and respiratory complications. We recently showed CHD-heterotaxy patients can have respiratory ciliary dysfunction (CD) similar to that associated with primary ciliary dyskinesia, including low nasal nitric oxide and abnormal ciliary motion. In this study, we investigated whether CHD-heterotaxy patients with CD may have worse postsurgical outcomes.nnnMETHODSnWe examined postsurgical outcome in 13 heterotaxy-CHD patients with CD (25 surgeries), compared with 14 heterotaxy-CHD patients without CD (27 surgeries). Outcome data were collected for each surgery, including respiratory complications, tracheostomy, use of inhaled β-agonists or nitric oxide, length of hospital stay, days on ventilator, and death.nnnRESULTSnThe CD versus the no-CD CHD cohorts had similar Risk Adjustment in Congenital Heart Surgery-1 risk categories, repair track, age at surgery, and follow-up evaluation times. Respiratory complications (76% vs 37%; Pxa0=xa0.006), need for tracheostomy (16% vs 0%; Pxa0=xa0.047), and use of inhaled β-agonists (64% vs 11%; Pxa0=xa0.0001) all were increased significantly in heterotaxy-CHD patients with CD. No significant differences were detected in postoperative hospital stay, days on mechanical ventilation, or surgical mortality. A trend toward increased mortality for the CD group beyond the postoperative period was observed (33% vs 0%; Pxa0=xa0.055) in patients younger than age 10 years.nnnCONCLUSIONSnOur findings showed that heterotaxy-CHD patients with CD may have increased risks for respiratory deficiencies. Overall, there was a trend toward increased mortality in CD patients with intermediate follow-up evaluation. Because β-agonists are known to increase ciliary beat frequency, presurgical screening for CD and perioperative treatment of CD patients with inhaled β-agonists may improve postoperative outcomes and survival.


PLOS Genetics | 2016

DNAH6 and Its Interactions with PCD Genes in Heterotaxy and Primary Ciliary Dyskinesia.

You Li; Hisato Yagi; Ezenwa O. Onuoha; Rama Rao Damerla; Richard Francis; Yoshiyuki Furutani; Muhammad Tariq; Stephen M. King; Gregory M. Hendricks; Cheng Cui; Dong Min Lee; Maliha Zahid; Iman Sami; Linda Leatherbury; Gregory J. Pazour; Stephanie M. Ware; Toshio Nakanishi; Elizabeth Goldmuntz; Michael Tsang; Cecilia W. Lo

Heterotaxy, a birth defect involving left-right patterning defects, and primary ciliary dyskinesia (PCD), a sinopulmonary disease with dyskinetic/immotile cilia in the airway are seemingly disparate diseases. However, they have an overlapping genetic etiology involving mutations in cilia genes, a reflection of the common requirement for motile cilia in left-right patterning and airway clearance. While PCD is a monogenic recessive disorder, heterotaxy has a more complex, largely non-monogenic etiology. In this study, we show mutations in the novel dynein gene DNAH6 can cause heterotaxy and ciliary dysfunction similar to PCD. We provide the first evidence that trans-heterozygous interactions between DNAH6 and other PCD genes potentially can cause heterotaxy. DNAH6 was initially identified as a candidate heterotaxy/PCD gene by filtering exome-sequencing data from 25 heterotaxy patients stratified by whether they have airway motile cilia defects. dnah6 morpholino knockdown in zebrafish disrupted motile cilia in Kupffer’s vesicle required for left-right patterning and caused heterotaxy with abnormal cardiac/gut looping. Similarly DNAH6 shRNA knockdown disrupted motile cilia in human and mouse respiratory epithelia. Notably a heterotaxy patient harboring heterozygous DNAH6 mutation was identified to also carry a rare heterozygous PCD-causing DNAI1 mutation, suggesting a DNAH6/DNAI1 trans-heterozygous interaction. Furthermore, sequencing of 149 additional heterotaxy patients showed 5 of 6 patients with heterozygous DNAH6 mutations also had heterozygous mutations in DNAH5 or other PCD genes. We functionally assayed for DNAH6/DNAH5 and DNAH6/DNAI1 trans-heterozygous interactions using subthreshold double-morpholino knockdown in zebrafish and showed this caused heterotaxy. Similarly, subthreshold siRNA knockdown of Dnah6 in heterozygous Dnah5 or Dnai1 mutant mouse respiratory epithelia disrupted motile cilia function. Together, these findings support an oligogenic disease model with broad relevance for further interrogating the genetic etiology of human ciliopathies.

Collaboration


Dive into the Richard Francis's collaboration.

Top Co-Authors

Avatar

Cecilia W. Lo

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar

Xiaoqin Liu

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Linda Leatherbury

Georgia Regents University

View shared research outputs
Top Co-Authors

Avatar

You Li

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar

Bishwanath Chatterjee

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Maliha Zahid

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar

Gregory J. Pazour

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar

William A. Devine

Boston Children's Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge