Network


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

Hotspot


Dive into the research topics where Stefan Heller is active.

Publication


Featured researches published by Stefan Heller.


Cell | 2000

Vanilloid Receptor–Related Osmotically Activated Channel (VR-OAC), a Candidate Vertebrate Osmoreceptor

Wolfgang Liedtke; Yong Choe; Marc A. Marti-Renom; Andrea M. Bell; Charlotte S. Denis; AndrejŠali; A. J. Hudspeth; Jeffrey M. Friedman; Stefan Heller

The detection of osmotic stimuli is essential for all organisms, yet few osmoreceptive proteins are known, none of them in vertebrates. By employing a candidate-gene approach based on genes encoding members of the TRP superfamily of ion channels, we cloned cDNAs encoding the vanilloid receptor-related osmotically activated channel (VR-OAC) from the rat, mouse, human, and chicken. This novel cation-selective channel is gated by exposure to hypotonicity within the physiological range. In the central nervous system, the channel is expressed in neurons of the circumventricular organs, neurosensory cells responsive to systemic osmotic pressure. The channel also occurs in other neurosensory cells, including inner-ear hair cells, sensory neurons, and Merkel cells.


Molecular Cell | 2002

A Unified Nomenclature for the Superfamily of TRP Cation Channels

Craig Montell; Lutz Birnbaumer; Veit Flockerzi; René J. M. Bindels; Elspeth A. Bruford; Michael J. Caterina; David E. Clapham; Christian Harteneck; Stefan Heller; David Julius; Itaru Kojima; Yasuo Mori; Reinhold Penner; Dirk Prawitt; Andrew M. Scharenberg; Günter Schultz; Nobuyoshi Shimizu; Michael X. Zhu

The TRP superfamily includes a diversity of non-voltage-gated cation channels that vary significantly in their selectivity and mode of activation. Nevertheless, members of the TRP superfamily share significant sequence homology and predicted structural similarities. Currently, most of the genes and proteins that comprise the TRP superfamily have multiple names and, in at least one instance, two distinct genes belonging to separate subfamilies have the same name. Moreover, there are many cases in which highly related proteins that belong to the same subfamily have unrelated names. Therefore, to minimize confusion, we propose a unified nomenclature for the TRP superfamily.The current effort to unify the TRP nomenclature focuses on three subfamilies (TRPC, TRPV, and TRPM) that bear significant similarities to the founding member of this superfamily, Drosophila TRP, and which include highly related members in worms, flies, mice, and humans (Table 1)(Table 1). Members of the three subfamilies contain six transmembrane segments, a pore loop separating the final two transmembrane segments, and similarity in the lengths of the cytoplasmic and extracellular loops. In addition, the charged residues in the S4 segment that appear to contribute to the voltage sensor in voltage-gated ion channels are not conserved. The TRP-Canonical (TRPC) subfamily (formerly short-TRPs or STRPs) is comprised of those proteins that are the most highly related to Drosophila TRP. The TRPV subfamily (formerly OTRPC), is so named based on the original designation, Vanilloid Receptor 1 (VR1), for the first mammalian member of this subfamily (now TRPV1). The name for the TRPM subfamily (formerly long-TRPs or LTRPs) is derived from the first letter of Melastatin, the former name (now TRPM1) of the founding member of this third subfamily of TRP-related proteins. Based on amino acid homologies, the mammalian members of these three subfamilies can be subdivided into several groups each (Table 2Table 2 and Figure 1Figure 1) .Table 1Number of TRP Genes in Worms (C. elegans), Flies (Drosophila melanogaster), Mice, and HumansSubfamilyWormsFliesMiceHumansTRPC3376aaTRPV5255TRPM4188aTRPC2 is a pseudogene and is not counted.Table 2Nomenclature of the Mammalian TRP SuperfamilyNameGroupFormer NamesAccession NumbersTRPC11TRP1CAA61447, AAA93252TRPC1TRPC22TRP2X89067, AAD17195, AAD17196, AAG29950, AAG29951, AAD31453,TRPC2CAA06964TRPC33TRP3AAC51653TRPC3TRPC44TRP4CAA68125, BAA23599TRPC4TRPC54TRP5AAC13550, CAA06911, CAA06912TRPC5TRPC63TRP6NP_038866TRPC6TRPC73TRP7AAD42069, NP_065122TRPC7TRPV11VR1AAC53398OTRPC1TRPV21VRL-1AAD26363, AAD26364, BAA78478OTRPC2GRCTRPV3 (not assigned)TRPV42OTRPC4AAG17543, AAG16127, AAG28027, AAG28028, AAG28029,VR-OACCAC20703TRP12VRL-2TRPV53ECaC1CAB40138CaT2TRPV63CaT1AAD47636ECaC2CAC20416CaT-LCAC20417TRPM11MelastatinAAC13683, AAC80000TRPM22TRPC7BAA34700LTRPC2TRPM31KIAA1616AA038185LTRPC3TRPM43TRPM4H18835LTRPC4TRPM53MTR1AAF26288LTRPC5TRPM64Chak2AF350881TRPM74TRP-PLIKAAF73131Chak1LTRPC7TRPM82TRP-p8AC005538Indicated are the suggested gene and protein names, the groups within each subfamily, the former names, and accession numbers.Figure 1Phylogenetic Tree of the TRP SuperfamilyThe tree, which was adapted from Clapham et al., 2001 (Nat. Rev. Neurosci. 2, 387–396), was calculated using the neighbor-joining method and human, rat, and mouse sequences.View Large Image | View Hi-Res Image | Download PowerPoint SlideThe numbering system for the mammalian TRPC, TRPV, and TRPM proteins takes into account the order of their discovery and, in as many cases as possible, the number that has already been assigned to the genes and proteins (Table 2)(Table 2). In the case of the TRPV proteins, the numbering system is also based in part on the groupings of the TRPV proteins. New members of each subfamily will maintain the same root name and, with the exception of TRPV3, will be assigned the next number in the sequence. Currently, TRPV3 is unassigned to maintain the TRPV1/ TRPV2 and TRPV5/TRPV6 groupings and so that the former OTRPC4 could be renamed TRPV4. The next TRPV protein will be designated TRPV3.We hope this new nomenclature will add clarity to the field and simplify the naming of new members of the TRP superfamily. We recommend that accession numbers be used whenever it is necessary to unambiguously specify a given variant resulting from alternative mRNA splicing. Finally, this nomenclature has been approved by the HUGO Gene Nomenclature Committee and we recommend that this system be used in all future publications concerning TRPC, TRPV, and TRPM subfamily members.


Nature Medicine | 2003

Pluripotent stem cells from the adult mouse inner ear

Huawei Li; Hong Liu; Stefan Heller

In mammals, the permanence of acquired hearing loss is mostly due to the incapacity of the cochlea to replace lost mechanoreceptor cells, or hair cells. In contrast, damaged vestibular organs can generate new hair cells, albeit in limited numbers. Here we show that the adult utricular sensory epithelium contains cells that display the characteristic features of stem cells. These inner ear stem cells have the capacity for self-renewal, and form spheres that express marker genes of the developing inner ear and the nervous system. Inner ear stem cells are pluripotent and can give rise to a variety of cell types in vitro and in vivo, including cells representative of ectodermal, endodermal and mesodermal lineages. Our observation that these stem cells are capable of differentiating into hair cell–like cells implies a possible use of such cells for the replacement of lost inner-ear sensory cells.


Nature Genetics | 1998

Mutations in a novel cochlear gene cause DFNA9, a human nonsyndromic deafness with vestibular dysfunction

Nahid G. Robertson; Leonard Lu; Stefan Heller; Saumil N. Merchant; Roland D. Eavey; Michael J. McKenna; Joseph B. Nadol; Richard T. Miyamoto; Frederick H. Linthicum; José Faibes Lubianca Neto; A.J. Hudspeth; Christine E. Seidman; Cynthia C. Morton; Jonathan G. Seidman

DFNA9 is an autosomal dominant, nonsyndromic, progressive sensorineural hearing loss with vestibular pathology. Here we report three missense mutations in human COCH (previously described as Coch5b2), a novel cochlear gene, in three unrelated kindreds with DFNA9. All three residues mutated in DFNA9 are conserved in mouse and chicken Coch, and are found in a region containing four conserved cysteines with homology to a domain in factor C, a lipopolysaccharide-binding coagulation factor in Limulus polyphemus. COCH message, found at high levels in human cochlear and vestibular organs, occurs in the chicken inner ear in the regions of the auditory and vestibular nerve fibres, the neural and abneural limbs adjacent to the cochlear sensory epithelium and the stroma of the crista ampullaris of the vestibular labyrinth. These areas correspond to human inner ear structures which show histopathological findings of acidophilic ground substance in DFNA9 patients.


Proceedings of the National Academy of Sciences of the United States of America | 2003

Generation of hair cells by stepwise differentiation of embryonic stem cells

Huawei Li; Graham Roblin; Hong Liu; Stefan Heller

The increase in life expectancy is accompanied by the growing burden of chronic diseases. Hearing loss is perhaps the most prevalent of all chronic diseases. In addition to age-related hearing loss, a substantial number of cases of audiological impairment are either congenital in nature or acquired during childhood. The permanence of hearing loss is mainly due to the inability of the cochlear sensory epithelium to replace lost mechanoreceptor cells, or hair cells. Generation of hair cells from a renewable source of progenitors that can be transplanted into damaged inner ears is a principal requirement for potential cell replacement therapy in this organ. Here, we present an experimental protocol that enables us to routinely create inner ear progenitors from murine embryonic stem cells in vitro. These progenitors express a comprehensive set of marker genes that define the developing inner ear, in particular the organs developing sensory patches. We further demonstrate that cells that express markers characteristic of hair cells differentiate from embryonic stem cell-derived progenitors. Finally, we show that these progenitors integrate into the developing inner ear at sites of epithelial injury and that integrated cells start expressing hair cell markers and display hair bundles when situated in cochlear or vestibular sensory epithelia in vivo.


Jaro-journal of The Association for Research in Otolaryngology | 2007

Differential Distribution of Stem Cells in the Auditory and Vestibular Organs of the Inner Ear

Kazuo Oshima; Christian Grimm; C. Eduardo Corrales; Pascal Senn; Rodrigo Martinez Monedero; Gwenaëlle S. G. Géléoc; Albert Edge; Jeffrey R. Holt; Stefan Heller

The adult mammalian cochlea lacks regenerative capacity, which is the main reason for the permanence of hearing loss. Vestibular organs, in contrast, replace a small number of lost hair cells. The reason for this difference is unknown. In this work we show isolation of sphere-forming stem cells from the early postnatal organ of Corti, vestibular sensory epithelia, the spiral ganglion, and the stria vascularis. Organ of Corti and vestibular sensory epithelial stem cells give rise to cells that express multiple hair cell markers and express functional ion channels reminiscent of nascent hair cells. Spiral ganglion stem cells display features of neural stem cells and can give rise to neurons and glial cell types. We found that the ability for sphere formation in the mouse cochlea decreases about 100-fold during the second and third postnatal weeks; this decrease is substantially faster than the reduction of stem cells in vestibular organs, which maintain their stem cell population also at older ages. Coincidentally, the relative expression of developmental and progenitor cell markers in the cochlea decreases during the first 3 postnatal weeks, which is in sharp contrast to the vestibular system, where expression of progenitor cell markers remains constant or even increases during this period. Our findings indicate that the lack of regenerative capacity in the adult mammalian cochlea is either a result of an early postnatal loss of stem cells or diminishment of stem cell features of maturing cochlear cells.


Pflügers Archiv: European Journal of Physiology | 2005

The mechanosensitive nature of TRPV channels

Roger G. O'Neil; Stefan Heller

Transient receptor potential vanilloid (TRPV) channels are widely expressed in both sensory and nonsensory cells. Whereas the channels display a broad diversity to activation by chemical and physical stimuli, activation by mechanical stimuli is common to many members of this group in both lower and higher organisms. Genetic screening in Caenorhabditis elegans has demonstrated an essential role for two TRPV channels in sensory neurons. OSM-9 and OCR-2, for example, are essential for both osmosensory and mechanosensory (nose-touch) behaviors. Likewise, two Drosophila TRPV channels, NAN and IAV, have been shown to be critical for hearing by the mechanosensitive chordotonal organs located in the fly’s antennae. The mechanosensitive nature of the channels appears to be conserved in higher organisms for some TRPV channels. Two vertebrate channels, TRPV2 and TRPV4, are sensitive to hypotonic cell swelling, shear stress/fluid flow (TRPV4), and membrane stretch (TRPV2). In the osmosensing neurons of the hypothalamus (circumventricular organs), TRPV4 appears to function as an osmoreceptor, or part of an osmoreceptor complex, in control of vasopressin release, whereas in inner ear hair cells and vascular baroreceptors a mechanosensory role is suggestive, but not demonstrated. Finally, in many nonsensory cells expressing TRPV4, such as vascular endothelial cells and renal tubular epithelial cells, the channel exhibits well-developed local mechanosensory transduction processes where both cell swelling and shear stress/fluid flow lead to channel activation. Hence, many TRPV channels, or combinations of TRPV channels, display a mechanosensitive nature that underlies multiple mechanosensitive processes from worms to mammals.


Cell | 2010

Mechanosensitive Hair Cell-like Cells from Embryonic and Induced Pluripotent Stem Cells

Kazuo Oshima; Kunyoo Shin; Marc Diensthuber; Anthony W. Peng; Anthony J. Ricci; Stefan Heller

Mechanosensitive sensory hair cells are the linchpin of our senses of hearing and balance. The inability of the mammalian inner ear to regenerate lost hair cells is the major reason for the permanence of hearing loss and certain balance disorders. Here, we present a stepwise guidance protocol starting with mouse embryonic stem and induced pluripotent stem cells, which were directed toward becoming ectoderm capable of responding to otic-inducing growth factors. The resulting otic progenitor cells were subjected to varying differentiation conditions, one of which promoted the organization of the cells into epithelial clusters displaying hair cell-like cells with stereociliary bundles. Bundle-bearing cells in these clusters responded to mechanical stimulation with currents that were reminiscent of immature hair cell transduction currents.


Neuron | 1997

Distribution of Ca2+-Activated K+ Channel Isoforms along the Tonotopic Gradient of the Chicken's Cochlea

Kevin P. Rosenblatt; Zhong-Ping Sun; Stefan Heller; A. J. Hudspeth

In some cochleae, the number and kinetic properties of Ca2+-activated K+ (KCa) channels partly determine the characteristic frequency of each hair cell and thus help establish a tonotopic map. In the chickens basilar papilla, we found numerous isoforms of KCa channels generated by alternative mRNA splicing at seven sites in a single gene, cSlo. In situ polymerase chain reactions demonstrated cSlo expression in hair cells and revealed differential distributions of KCa channel isoforms along the basilar papilla. Analysis of single hair cells by the reverse transcription polymerase chain reaction confirmed the differential expression of channel variants. Heterologously expressed cSlo variants differed in their sensitivities to Ca2+ and voltage, suggesting that the distinct spatial distributions of cSlo variants help determine the tonotopic map.


Journal of Biological Chemistry | 2006

PACSINs Bind to the TRPV4 Cation Channel PACSIN 3 MODULATES THE SUBCELLULAR LOCALIZATION OF TRPV4

Math P. Cuajungco; Christian Grimm; Kazuo Oshima; Dieter D'hoedt; Bernd Nilius; Arjen R. Mensenkamp; René J. M. Bindels; Markus Plomann; Stefan Heller

TRPV4 is a cation channel that responds to a variety of stimuli including mechanical forces, temperature, and ligand binding. We set out to identify TRPV4-interacting proteins by performing yeast two-hybrid screens, and we isolated with the avian TRPV4 amino terminus the chicken orthologues of mammalian PACSINs 1 and 3. The PACSINs are a protein family consisting of three members that have been implicated in synaptic vesicular membrane trafficking and regulation of dynamin-mediated endocytotic processes. In biochemical interaction assays we found that all three murine PACSIN isoforms can bind to the amino terminus of rodent TRPV4. No member of the PACSIN protein family was able to biochemically interact with TRPV1 and TRPV2. Co-expression of PACSIN 3, but not PACSINs 1 and 2, shifted the ratio of plasma membrane-associated versus cytosolic TRPV4 toward an apparent increase of plasma membrane-associated TRPV4 protein. A similar shift was also observable when we blocked dynamin-mediated endocytotic processes, suggesting that PACSIN 3 specifically affects the endocytosis of TRPV4, thereby modulating the subcellular localization of the ion channel. Mutational analysis shows that the interaction of the two proteins requires both a TRPV4-specific proline-rich domain upstream of the ankyrin repeats of the channel and the carboxyl-terminal Src homology 3 domain of PACSIN 3. Such a functional interaction could be important in cell types that show distribution of both proteins to the same subcellular regions such as renal tubule cells where the proteins are associated with the luminal plasma membrane.

Collaboration


Dive into the Stefan Heller's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

C. Eduardo Corrales

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

A. J. Hudspeth

Howard Hughes Medical Institute

View shared research outputs
Top Co-Authors

Avatar

Hong Liu

Massachusetts Eye and Ear Infirmary

View shared research outputs
Top Co-Authors

Avatar

Math P. Cuajungco

California State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hermann Rohrer

Goethe University Frankfurt

View shared research outputs
Researchain Logo
Decentralizing Knowledge